51
|
Sugihara HY, Okamoto R, Mizutani T. Intestinal organoids: The path towards clinical application. Eur J Cell Biol 2025; 104:151474. [PMID: 39740324 DOI: 10.1016/j.ejcb.2024.151474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 01/02/2025] Open
Abstract
Organoids have revolutionized the whole field of biology with their ability to model complex three-dimensional human organs in vitro. Intestinal organoids were especially consequential as the first successful long-term culture of intestinal stem cells, which raised hopes for translational medical applications. Despite significant contributions to basic research, challenges remain to develop intestinal organoids into clinical tools for diagnosis, prognosis, and therapy. In this review, we outline the current state of translational research involving adult stem cell and pluripotent stem cell derived intestinal organoids, highlighting the advances and limitations in disease modeling, drug-screening, personalized medicine, and stem cell therapy. Preclinical studies have demonstrated a remarkable functional recapitulation of infectious and genetic diseases, and there is mounting evidence for the reliability of intestinal organoids as a patient-specific avatar. Breakthroughs now allow the generation of structurally and cellularly complex intestinal models to better capture a wider range of intestinal pathophysiology. As the field develops and evolves, there is a need for standardized frameworks for generation, culture, storage, and analysis of intestinal organoids to ensure reproducibility, comparability, and interpretability of these preclinical and clinical studies to ultimately enable clinical translation.
Collapse
Affiliation(s)
- Hady Yuki Sugihara
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Tomohiro Mizutani
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
52
|
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
|
53
|
Choi Y, Na D, Yoon G, Kim J, Min S, Yi H, Cho S, Cho JH, Lee C, Jang J. Prediction of Patient Drug Response via 3D Bioprinted Gastric Cancer Model Utilized Patient-Derived Tissue Laden Tissue-Specific Bioink. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411769. [PMID: 39748450 PMCID: PMC11905052 DOI: 10.1002/advs.202411769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/20/2024] [Indexed: 01/04/2025]
Abstract
Despite significant research progress, tumor heterogeneity remains elusive, and its complexity poses a barrier to anticancer drug discovery and cancer treatment. Response to the same drug varies across patients, and the timing of treatment is an important factor in determining prognosis. Therefore, development of patient-specific preclinical models that can predict a patient's drug response within a short period is imperative. In this study, a printed gastric cancer (pGC) model is developed for preclinical chemotherapy using extrusion-based 3D bioprinting technology and tissue-specific bioinks containing patient-derived tumor chunks. The pGC model retained the original tumor characteristics and enabled rapid drug evaluation within 2 weeks of its isolation from the patient. In fact, it is confirmed that the drug response-related gene profile of pGC tissues co-cultured with human gastric fibroblasts (hGaFibro) is similar to that of patient tissues. This suggested that the application of the pGC model can potentially overcome the challenges associated with accurate drug evaluation in preclinical models (e.g., patient-derived xenografts) owing to the deficiency of stromal cells derived from the patient. Consequently, the pGC model manifested a remarkable similarity with patients in terms of response to chemotherapy and prognostic predictability. Hence, it is considered a promising preclinical tool for personalized and precise treatments.
Collapse
Affiliation(s)
- Yoo‐mi Choi
- Center for 3D Organ Printing and Stem cells (COPS)Pohang University of Science and Technology (POSTECH)Pohang37666Republic of Korea
| | - Deukchae Na
- Ewha Institute of Convergence MedicineEwha Womans University Mokdong HospitalSeoul07985Republic of Korea
| | - Goeun Yoon
- Department of Mechanical EngineeringPohang University of Science and Technology (POSTECH)Pohang37666Republic of Korea
| | - Jisoo Kim
- School of Interdisciplinary Bioscience and BioengineeringPohang University of Science and Technology (POSTECH)Pohang37666Republic of Korea
| | - Seoyeon Min
- Ewha Institute of Convergence MedicineEwha Womans University Mokdong HospitalSeoul07985Republic of Korea
| | - Hee‐Gyeong Yi
- Department of Rural and Biosystems EngineeringChonnam National UniversityGwangju61186Republic of Korea
| | - Soo‐Jeong Cho
- Department of Internal MedicineLiver Research InstituteSeoul National University HospitalSeoul03080Republic of Korea
| | - Jae Hee Cho
- Department of Internal MedicineGangnam Severance HospitalYonsei University College of MedicineSeoul06273Republic of Korea
| | - Charles Lee
- Ewha Institute of Convergence MedicineEwha Womans University Mokdong HospitalSeoul07985Republic of Korea
- The Jackson Laboratory for Genomic MedicineFarmingtonCT06032USA
| | - Jinah Jang
- Center for 3D Organ Printing and Stem cells (COPS)Pohang University of Science and Technology (POSTECH)Pohang37666Republic of Korea
- Department of Mechanical EngineeringPohang University of Science and Technology (POSTECH)Pohang37666Republic of Korea
- School of Interdisciplinary Bioscience and BioengineeringPohang University of Science and Technology (POSTECH)Pohang37666Republic of Korea
- Department of Convergence IT EngineeringPohang University of Science and Technology (POSTECH)Pohang37666Republic of Korea
- Institute for Convergence Research and Education in Advanced TechnologyYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|
54
|
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]
|
55
|
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
|
56
|
Klompstra TM, Yoon KJ, Koo BK. Evolution of organoid genetics. Eur J Cell Biol 2025; 104:151481. [PMID: 40056574 DOI: 10.1016/j.ejcb.2025.151481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/01/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025] Open
Abstract
Organoids have revolutionized in vitro research by offering three-dimensional, multicellular systems that recapitulate the structure, function, and genetics of human tissues. Initially developed from both pluripotent stem cells (PSCs) and adult stem cells (AdSCs), organoids have expanded to model nearly every major human organ, significantly advancing developmental biology, disease modeling, and therapeutic screening. This review highlights the progression of organoid technologies, emphasizing the integration of genetic tools, including CRISPR-Cas9, prime editing, and lineage tracing. These advancements have facilitated precise modeling of human-specific pathologies and drug responses, often surpassing traditional 2D cultures and animal models in accuracy. Emerging technologies, such as organoid fusion, xenografting, and optogenetics, are expected to further enhance our understanding of cellular interactions and microenvironmental dynamics. As organoid complexity and genetic engineering methods continue to evolve, they will become increasingly indispensable for personalized medicine and translational research, bridging gaps between in vitro and in vivo systems.
Collapse
Affiliation(s)
- Thomas M Klompstra
- Center for Genome Engineering, Institute for Basic Sciences (IBS), Republic of Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Republic of Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Republic of Korea; Graduate School of Stem Cell and Regenerative Biology, KAIST, Daejeon 34141, Republic of Korea; KAIST Stem Cell Center, KAIST, Daejeon 34141, Republic of Korea
| | - Bon-Kyoung Koo
- Center for Genome Engineering, Institute for Basic Sciences (IBS), Republic of Korea; Graduate School of Stem Cell and Regenerative Biology, KAIST, Daejeon 34141, Republic of Korea; Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Republic of Korea.
| |
Collapse
|
57
|
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] [Download PDF] [Figures] [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
|
58
|
Wu T, Huang J, Zhang X, Ma F, Yu S, Liu Y, Xu Y, Wu S, Finko AV, Li Z, Feng L, Bian J. Rational Design of a Potent, Selective, and Metabolically Stable CDK9 Inhibitor to Counteract Osimertinib Resistance through Mcl-1 Suppression and Enhanced BRD4 Co-Targeting. J Med Chem 2025; 68:4929-4950. [PMID: 39947929 DOI: 10.1021/acs.jmedchem.4c03168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Overcoming osimertinib resistance in NSCLC treatment remains a significant clinical challenge. CDK9 has emerged as a promising target due to its critical role in sustaining oncogenic transcriptional programs, particularly via Mcl-1 regulation. Herein, we report the structure-guided optimization of a previously identified CDK9 inhibitor (Z11), resulting in the discovery of T7, a potent, selective, and metabolically stable candidate (IC50 = 1.2 nM). T7 effectively suppressed cell proliferation, reduced colony formation, and induced apoptosis in Osimertinib-resistant NSCLC cells by downregulating Mcl-1. Furthermore, T7 significantly inhibited the growth of resistant organoids and demonstrated marked antitumor efficacy in a xenograft model. Notably, combining T7 with the BRD4 inhibitor JQ1 further enhanced antitumor activity both in vitro and in vivo, revealing a complementary therapeutic strategy. These findings identify T7 as a promising next-generation CDK9 inhibitor for addressing Osimertinib resistance in NSCLC and underscore the potential of transcriptional cotargeting approaches to improve clinical outcomes.
Collapse
Affiliation(s)
- Tizhi Wu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jiali Huang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xiankang Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Feihai Ma
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Shijing Yu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yifang Liu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yifan Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Shiqi Wu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Alexander V Finko
- Department of Chemistry, Lomonosov Moscow State University (MSU), Moscow 119991, Russia
| | - Zhiyu Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Li Feng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Jinlei Bian
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| |
Collapse
|
59
|
Ogawa H, Yoshida K, Hasegawa S, Wada H, Yasui M, Tahara H. Significance of mouse xenograft tumor model using patient-derived cancer organoids for clinical drug development. Front Oncol 2025; 15:1485886. [PMID: 40078183 PMCID: PMC11896854 DOI: 10.3389/fonc.2025.1485886] [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: 08/25/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
Background In vitro and in vivo preclinical examinations of cancer cell lines are performed to determine the effectiveness of new drugs before initiating clinical trials. However, there is often a significant disparity between the promising results observed in preclinical evaluations and actual outcomes in clinical trials. Therefore, we hypothesized that this inconsistency might be due to the differences between the characteristics of cell lines and actual cancers in patients. Therefore, we screened drugs for bile duct cancer to test our hypotheses. Methods We established patient-derived cancer organoids (PDCOs) from the surgical samples of patients with bile duct cancer and conducted multiple in vitro drug screening tests. Results We identified proteasome inhibitors (Bortezomib and Carfilzomib) as promising drugs in the screening. Bortezomib has demonstrated a significant antitumor effect on bile duct cancer cell-derived xenografts, as previously reported in preclinical trials. However, although Bortezomib showed significant proliferation inhibition in PDCOs in three-dimensional culture in vitro, it did not exhibit significant anti-tumor effects in mouse xenograft tumor models using our PDCOs. Bile duct cancer cell-line-derived xenografts are characterized by structurally uniform, irregular glandular structures surrounded by simple and sparse stromal components. However, organoid-derived xenografts exhibit a spectrum of differentiation levels within irregular glandular structures and consist of a complex and rich stromal microenvironment similar to those observed in surgical specimens. Conclusion These findings suggest that in vivo studies using PDCO xenograft tumor models may be more suitable than conventional mouse tumor models for determining the clinical development of drugs.
Collapse
Affiliation(s)
- Hisataka Ogawa
- Nitto joint Research Department for Nucleic Acid Medicine, Research Center, Osaka International Cancer Institute, Osaka, Japan
| | - Keiichi Yoshida
- Next-generation Precision Medicine Research Center, Osaka International Cancer Institute, Osaka, Japan
| | - Shinichiro Hasegawa
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Hiroshi Wada
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Masayoshi Yasui
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Hideaki Tahara
- Department of Cancer Drug Discovery and Development, Research Center, Osaka International Cancer Institute, Osaka, Japan
- Center for Clinical Research, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
60
|
Artegiani B, Hendriks D. Organoids from pluripotent stem cells and human tissues: When two cultures meet each other. Dev Cell 2025; 60:493-511. [PMID: 39999776 DOI: 10.1016/j.devcel.2025.01.005] [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: 03/11/2024] [Revised: 06/13/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
Human organoids are a widely used tool in cell biology to study homeostatic processes, disease, and development. The term organoids covers a plethora of model systems from different cellular origins that each have unique features and applications but bring their own challenges. This review discusses the basic principles underlying organoids generated from pluripotent stem cells (PSCs) as well as those derived from tissue stem cells (TSCs). We consider how well PSC- and TSC-organoids mimic the different intended organs in terms of cellular complexity, maturity, functionality, and the ongoing efforts to constitute predictive complex models of in vivo situations. We discuss the advantages and limitations associated with each system to answer different biological questions including in the field of cancer and developmental biology, and with respect to implementing emerging advanced technologies, such as (spatial) -omics analyses, CRISPR screens, and high-content imaging screens. We postulate how the two fields may move forward together, integrating advantages of one to the other.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
61
|
Mahmoud A, Choi PH, Sukhwa C, Pintar J, Walch H, Zhao N, Bermeo J, Chung S, Raghavan M, Bapat S, Jiang Q, Karagkounis G, Meredith J, Giarrizzo M, Firat C, Cercek A, Foote MB, Schultz N, Chatila WK, Nash GM, Shia J, Sanchez-Vega F, Larson S, Dar AC, Rosen N, Ganesh K. Paired primary-metastasis patient-derived organoids and mouse models identify phenotypic evolution and druggable dependencies of peritoneal metastasis from appendiceal cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638725. [PMID: 40027618 PMCID: PMC11870485 DOI: 10.1101/2025.02.17.638725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Peritoneal carcinomatosis is a common yet deadly manifestation of gastrointestinal cancers, with few effective treatments. To identify targetable determinants of peritoneal metastasis, we focused on appendiceal adenocarcinoma (AC), a gastrointestinal cancer that metastasizes almost exclusively to the peritoneum. Current treatments are extrapolated from colorectal cancer (CRC), yet AC has distinct genomic alterations, mucinous morphology and peritoneum restricted metastatic pattern. Further, no stable preclinical models of AC exist, limiting drug discovery and representing an unmet clinical need. We establish a first-in-class stable biobank of 16 long-term cultured AC patient-derived organoids (PDOs), including 3 matched, simultaneously resected primary AC-peritoneal carcinomatosis (AC-PC) pairs. By enriching for cancer cells, AC PDOs enable accurate genomic characterization relative to paucicellular AC tissue. We establish an organoid orthotopic intraperitoneal xenograft model that recapitulates diffuse peritoneal carcinomatosis and show that PC-organoids retain increased metastatic capacity, decreased growth factor dependency and sensitivity to standard of care chemotherapy relative to matched primary AC organoids. Single cell profiling of AC-PC pairs reveals dedifferentiation from mucinous differentiated states in primary AC into intestinal stem cell and fetal progenitor states in AC-PC, with upregulation of oncogenic signaling pathways. Through hypothesis-driven drug testing, we identify KRASMULTI-ON inhibitor RMC-7977 and Wnt-targeting tyrosine kinase inhibitor WNTinib as novel, clinically actionable strategies to target AC-PC more effectively.
Collapse
Affiliation(s)
- Ahmed Mahmoud
- Pharmacology Program, Weill Cornell Graduate School, New York, NY, USA
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Philip H. Choi
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christine Sukhwa
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jura Pintar
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Henry Walch
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nan Zhao
- Program in Chemical Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan Bermeo
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sebastian Chung
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Manisha Raghavan
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Program in Chemical Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samhita Bapat
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Qingwen Jiang
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Georgios Karagkounis
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Julia Meredith
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Giarrizzo
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Canan Firat
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Andrea Cercek
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael B. Foote
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Walid K. Chatila
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Garrett M. Nash
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Francisco Sanchez-Vega
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Steven Larson
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Arvin C. Dar
- Program in Chemical Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neal Rosen
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karuna Ganesh
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
62
|
Liu J, Cheng P, Xu C, Pu K. Molecular probes for in vivo optical imaging of immune cells. Nat Biomed Eng 2025:10.1038/s41551-024-01275-7. [PMID: 39984703 DOI: 10.1038/s41551-024-01275-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/23/2024] [Indexed: 02/23/2025]
Abstract
Advancing the understanding of the various roles and components of the immune system requires sophisticated methods and technology for the detection of immune cells in their natural states. Recent advancements in the development of molecular probes for optical imaging have paved the way for non-invasive visualization and real-time monitoring of immune responses and functions. Here we discuss recent progress in the development of molecular probes for the selective imaging of specific immune cells. We emphasize the design principles of the probes and their comparative performance when using various optical modalities across disease contexts. We highlight molecular probes for imaging tumour-infiltrating immune cells, and their applications in drug screening and in the prediction of therapeutic outcomes of cancer immunotherapies. We also discuss the use of these probes in visualizing immune cells in atherosclerosis, lung inflammation, allograft rejection and other immune-related conditions, and the translational opportunities and challenges of using optical molecular probes for further understanding of the immune system and disease diagnosis and prognosis.
Collapse
Affiliation(s)
- Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
63
|
Büning A, Reckzeh E. Opportunities of patient-derived organoids in drug development. Br J Pharmacol 2025. [PMID: 39978784 DOI: 10.1111/bph.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/02/2025] [Accepted: 02/01/2025] [Indexed: 02/22/2025] Open
Abstract
Various model systems are utilised during drug development starting from basic research, moving to preclinical research and development for clinical applications in order to identify new drugs to improve human health. However, there are characteristics of humans that are not captured by established models. Such models include homogeneous two-dimensional (2D) cell lines, which lack cellular heterogeneity and physiological relevance, and species differences of animal models. Organoids can mitigate these differences by providing more physiologically relevant three-dimensional (3D) cell models that resemble the molecular state in healthy and pathological tissue. This review presents exemplary approaches using patient-derived organoids (PDOs) that have been developed and the new opportunities that are evolving in drug development with a focus on patient adult stem cell (ASC)-derived organoids. These demonstrate the potential of PDOs used alongside established cell and animal models to improve drug development from basic research to clinical applications such as personalised medicine.
Collapse
Affiliation(s)
- Antonia Büning
- Transdisciplinary Research Area 'Life and Health', LIMES Institute, University of Bonn, Bonn, Germany
| | - Elena Reckzeh
- Transdisciplinary Research Area 'Life and Health', LIMES Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
64
|
Mallya D, Gadre MA, Varadharajan S, Vasanthan KS. 3D bioprinting for the construction of drug testing models-development strategies and regulatory concerns. Front Bioeng Biotechnol 2025; 13:1457872. [PMID: 40028291 PMCID: PMC11868281 DOI: 10.3389/fbioe.2025.1457872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025] Open
Abstract
A drug to be successfully launched in the market requires a significant amount of capital, resources and time, where the unsuccessful results in the last stages lead to catastrophic failure for discovering drugs. This is the very reason which calls for the invention of innovative models that can closely mimic the human in vivo model for producing reliable results. Throughout the innovation line, there has been improvement in the rationale in silico designing but yet there is requirement for in vitro-in vivo correlations. During the evolving of the drug testing models, the 3D models produced by different methods have been proven to produce better results than the traditional 2D models. However, the in vitro fabrications of live tissues are still bottleneck in realizing their complete potential. There is an urgent need for the development of single, standard and simplified in vitro 3D tissue models that can be reliable for investigating the biological and pathological aspects of drug discovery, which is yet to be achieved. The existing pre-clinical models have considerable drawbacks despite being the gold standard in pre-clinical research. The major drawback being the interspecies differences and low reliability on the generated results. This gap could be overcome by the fabrication of bioengineered human disease models for drug screening. The advancement in the fabrication of 3D models will provide a valuable tool in screening drugs at different stages as they are one step closer to bio-mimic human tissues. In this review, we have discussed on the evolution of preclinical studies, and different models, including mini tissues, spheroids, organoids, bioengineered three dimensional models and organs on chips. Furthermore, we provide details of different disease models fabricated across various organs and their applications. In addition to this, the review also focuses on the limitations and the current prospects of the role of three dimensionally bioprinted models in drug screening and development.
Collapse
Affiliation(s)
- Divya Mallya
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mrunmayi Ashish Gadre
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - S. Varadharajan
- Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kirthanashri S. Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
65
|
Riendeau JM, Gillette AA, Guzman EC, Cruz MC, Kralovec A, Udgata S, Schmitz A, Deming DA, Cimini BA, Skala MC. Cellpose as a reliable method for single-cell segmentation of autofluorescence microscopy images. Sci Rep 2025; 15:5548. [PMID: 39952935 PMCID: PMC11828867 DOI: 10.1038/s41598-024-82639-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 12/06/2024] [Indexed: 02/17/2025] Open
Abstract
Autofluorescence microscopy uses intrinsic sources of molecular contrast to provide cellular-level information without extrinsic labels. However, traditional cell segmentation tools are often optimized for high signal-to-noise ratio (SNR) images, such as fluorescently labeled cells, and unsurprisingly perform poorly on low SNR autofluorescence images. Therefore, new cell segmentation tools are needed for autofluorescence microscopy. Cellpose is a deep learning network that is generalizable across diverse cell microscopy images and automatically segments single cells to improve throughput and reduce inter-human biases. This study aims to validate Cellpose for autofluorescence imaging, specifically using multiphoton intensity images of NAD(P)H. Manually segmented nuclear masks of NAD(P)H images were used to train a new autofluorescence-trained model (ATM) in Cellpose for nuclear segmentation of NAD(P)H intensity images. These models were applied to PANC-1 cells treated with metabolic inhibitors and patient-derived cancer organoids (9 patients) treated with chemotherapies. These datasets include co-registered fluorescence lifetime imaging microscopy (FLIM) of NAD(P)H and FAD, so fluorescence decay parameters and the optical redox ratio (ORR) were compared between masks generated by the new ATM and manual segmentation. The Dice score between repeated manually segmented masks was significantly lower than that of repeated ATM masks (p < 0.0001) indicating greater reproducibility between ATM masks. There was also a high correlation (R2 > 0.9) between ATM and manually segmented masks for the ORR, mean NAD(P)H lifetime, and mean FAD lifetime across 2D and 3D cell culture treatment conditions. Masks generated from ATM and manual segmentation also maintain similar means, variances, and effect sizes between treatments for the ORR and FLIM parameters. Overall, the Cellpose ATM provides a fast, reliable, reproducible, and accurate method to segment single cells in autofluorescence microscopy images such that functional changes in cells are accurately captured in both 2D and 3D culture.
Collapse
Affiliation(s)
- Jeremiah M Riendeau
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | | | | | - Mario Costa Cruz
- Broad Institute of Harvard and MIT, Imaging Platform, Cambridge, MA, USA
| | | | - Shirsa Udgata
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Alexa Schmitz
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Dustin A Deming
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Beth A Cimini
- Broad Institute of Harvard and MIT, Imaging Platform, Cambridge, MA, USA
| | - Melissa C Skala
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.
- Morgridge Institute for Research, Madison, WI, USA.
| |
Collapse
|
66
|
Ren H, Wang YJ, Wang XY, Li X, Han Z, Zhang G, Gu L, Bai M, Yao GD, Liu Q, Song SJ. Design of ROS-Triggered Sesquiterpene Lactone SC Prodrugs as TrxR1 Covalent Inhibitors for the Treatment of Non-Small Cell Lung Cancer. J Med Chem 2025; 68:3088-3122. [PMID: 39869029 DOI: 10.1021/acs.jmedchem.4c02334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Thioredoxin reductase 1 (TrxR1) is an important therapeutic target for nonsmall cell lung cancer (NSCLC) treatment due to its overexpression in NSCLC cells. In this work, to address the deficiency that sesquiterpene lactone containing α-methylene-γ-lactone moiety was rapidly metabolized by endogenous nucleophiles, series of novel thioether derivatives were designed and synthesized based on a reactive oxygen species (ROS)-triggered prodrug strategy. Among them, prodrug 5u exhibited potent cytotoxicity against NSCLC cells and better release rates in response to ROS. The active compound 6a released from 5u covalently binds to Cys475 and Sec498 sites on TrxR1, resulting in inhibition on TrxR1 activity, which led to redox homeostasis disorder, and caused apoptosis and ferroptosis. Moreover, prodrug 5u exhibited significant antitumor efficiency in nude mice and NSCLC organoids. Our results deliver ROS-triggered prodrug 5u as a novel TrxR1 inhibitor for the treatment of NSCLC and provide a promising strategy of ROS-activated prodrug for covalent compounds in cancer therapy.
Collapse
Affiliation(s)
- Hui Ren
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yu-Jue Wang
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xin-Ye Wang
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xiangyun Li
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zheng Han
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Guxue Zhang
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Liwei Gu
- Institute of Chinese Materia Medica, Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ming Bai
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Guo-Dong Yao
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Qingbo Liu
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| |
Collapse
|
67
|
Tao W, Sun Q, Xu B, Wang R. Towards the Prediction of Responses to Cancer Immunotherapy: A Multi-Omics Review. Life (Basel) 2025; 15:283. [PMID: 40003691 PMCID: PMC11856636 DOI: 10.3390/life15020283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor treatment has undergone revolutionary changes with the development of immunotherapy, especially immune checkpoint inhibitors. Because not all patients respond positively to immune therapeutic agents, and severe immune-related adverse events (irAEs) are frequently observed, the development of the biomarkers evaluating the response of a patient is key for the application of immunotherapy in a wider range. Recently, various multi-omics features measured by high-throughput technologies, such as tumor mutation burden (TMB), gene expression profiles, and DNA methylation profiles, have been proved to be sensitive and accurate predictors of the response to immunotherapy. A large number of predictive models based on these features, utilizing traditional machine learning or deep learning frameworks, have also been proposed. In this review, we aim to cover recent advances in predicting tumor immunotherapy response using multi-omics features. These include new measurements, research cohorts, data sources, and predictive models. Key findings emphasize the importance of TMB, neoantigens, MSI, and mutational signatures in predicting ICI responses. The integration of bulk and single-cell RNA sequencing has enhanced our understanding of the tumor immune microenvironment and enabled the identification of predictive biomarkers like PD-L1 and IFN-γ signatures. Public datasets and machine learning models have also improved predictive tools. However, challenges remain, such as the need for large and diverse clinical datasets, standardization of multi-omics data, and model interpretability. Future research will require collaboration among researchers, clinicians, and data scientists to address these issues and enhance cancer immunotherapy precision.
Collapse
Affiliation(s)
- Weichu Tao
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (W.T.); (Q.S.)
| | - Qian Sun
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (W.T.); (Q.S.)
| | - Bingxiang Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (W.T.); (Q.S.)
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (W.T.); (Q.S.)
| |
Collapse
|
68
|
Wu Y, Sha Y, Guo X, Gao L, Huang J, Liu SB. Organoid models: applications and research advances in colorectal cancer. Front Oncol 2025; 15:1432506. [PMID: 39990692 PMCID: PMC11842244 DOI: 10.3389/fonc.2025.1432506] [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: 05/14/2024] [Accepted: 01/20/2025] [Indexed: 02/25/2025] Open
Abstract
This review summarizes the applications and research progress of organoid models in colorectal cancer research. First, the high incidence and mortality rates of colorectal cancer are introduced, emphasizing the importance of organoids as a research model. Second, this review provides a detailed introduction to the concept, biological properties, and applications of organoids, including their strengths in mimicking the structural and functional aspects of organs. This article further analyzes the applications of adult stem cell-derived and pluripotent stem cell-derived organoids in colorectal cancer research and discusses advancements in organoids for basic research, drug research and development, personalized treatment evaluation and prediction, and regenerative medicine. Finally, this review summarizes the prospects for applying organoid technology in colorectal cancer research, emphasizing its significant value in improving patient survival rates. In conclusion, this review systematically explains the applications of organoids in colorectal cancer research, highlighting their tremendous potential and promising prospects in basic research, drug research and development, personalized treatment evaluation and prediction, and regenerative medicine.
Collapse
Affiliation(s)
- Yijie Wu
- College of Life Science, North China University of Science and Technology, Tangshan, China
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Suzhou, China
| | - Yu Sha
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Suzhou, China
| | - Xingpo Guo
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ling Gao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Huang
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Suzhou, China
| | - Song-Bai Liu
- College of Life Science, North China University of Science and Technology, Tangshan, China
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Suzhou, China
| |
Collapse
|
69
|
Logun M, Wang X, Sun Y, Bagley SJ, Li N, Desai A, Zhang DY, Nasrallah MP, Pai ELL, Oner BS, Plesa G, Siegel D, Binder ZA, Ming GL, Song H, O'Rourke DM. Patient-derived glioblastoma organoids as real-time avatars for assessing responses to clinical CAR-T cell therapy. Cell Stem Cell 2025; 32:181-190.e4. [PMID: 39657679 PMCID: PMC11808387 DOI: 10.1016/j.stem.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 08/19/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024]
Abstract
Patient-derived tumor organoids have been leveraged for disease modeling and preclinical studies but rarely applied in real time to aid with interpretation of patient treatment responses in clinics. We recently demonstrated early efficacy signals in a first-in-human, phase 1 study of dual-targeting chimeric antigen receptor (CAR)-T cells (EGFR-IL13Rα2 CAR-T cells) in patients with recurrent glioblastoma. Here, we analyzed six sets of patient-derived glioblastoma organoids (GBOs) treated concurrently with the same autologous CAR-T cell products as patients in our phase 1 study. We found that CAR-T cell treatment led to target antigen reduction and cytolysis of tumor cells in GBOs, the degree of which correlated with CAR-T cell engraftment detected in patients' cerebrospinal fluid (CSF). Furthermore, cytokine release patterns in GBOs mirrored those in patient CSF samples over time. Our findings highlight a unique trial design and GBOs as a valuable platform for real-time assessment of CAR-T cell bioactivity and insights into immunotherapy efficacy.
Collapse
Affiliation(s)
- Meghan Logun
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Xin Wang
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yusha Sun
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Stephen J Bagley
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nannan Li
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arati Desai
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel Y Zhang
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - MacLean P Nasrallah
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Emily Ling-Lin Pai
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bike Su Oner
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Donald Siegel
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zev A Binder
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Hongjun Song
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Donald M O'Rourke
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
70
|
Zheng M, Qu J, Xiang D, Xing L. Organoids in lung cancer brain metastasis: Foundational research, clinical translation, and prospective outlooks. Biochim Biophys Acta Rev Cancer 2025; 1880:189235. [PMID: 39647672 DOI: 10.1016/j.bbcan.2024.189235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 12/01/2024] [Accepted: 12/01/2024] [Indexed: 12/10/2024]
Abstract
Brain metastasis stands as a leading contributor to mortality in lung cancer patients, yet the intricate mechanism underlying this phenomenon remains elusive. This underscores the need for robust preclinical models and effective treatment strategies. Emerging as viable in vitro models that closely replicate actual tumors, three-dimensional culture systems, particularly organoids derived from non-malignant cells or cancer organoids, have emerged as promising avenues. This review delves into the forefronts of fundamental research and clinical applications focused on lung cancer brain metastasis-derived organoids, highlighting current challenges and delineating prospects. These studies offer tremendous potential for clinical application despite being in nascent status.
Collapse
Affiliation(s)
- Mei Zheng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Jialin Qu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Dongxi Xiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China; Department of Biliary-Pancreatic Surgery, the Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, China.
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China.
| |
Collapse
|
71
|
Dong Y, Li J, Dai Y, Zhang X, Jiang X, Wang T, Zhao B, Liu W, Sun H, Du P, Qin L, Jiao Z. A novel nanocarrier based on natural polyphenols enhancing gemcitabine sensitization ability for improved pancreatic cancer therapy efficiency. Mater Today Bio 2025; 30:101463. [PMID: 39866791 PMCID: PMC11764724 DOI: 10.1016/j.mtbio.2025.101463] [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: 11/07/2024] [Revised: 01/04/2025] [Accepted: 01/05/2025] [Indexed: 01/28/2025] Open
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy with rapid progression and poor prognosis. Despite the widespread use of gemcitabine (Gem)-based chemotherapy as the first-line treatment for PC, its efficacy is often compromised by significant drug resistance. 1,2,3,4,6-Pentagaloyl glucose (PGG), a natural polyphenol, has demonstrated potential in sensitizing PC cells to Gem. However, its clinical application is limited by poor water solubility and bioavailability. In this study, we developed a novel PGG-based nanocarrier (FP) using a straightforward, one-step self-assembly method with Pluronic F127 and PGG. Our results showed that FP induced DNA damage and immunogenic cell death (ICD) in both in vitro cell experiments and patient-derived organoid models, exhibiting potent anti-tumor effects. Furthermore, in mouse KPC and PDX models, FP, when combined with Gem, showed enhanced Gem sensitization compared to pure PGG, largely due to increased DNA damage and ICD induction. These findings demonstrate the potential of FP to improve the stability and utilization of PGG as effective Gem sensitizers in the treatment of pancreatic cancer, providing a promising pathway for clinical application and translational research.
Collapse
Affiliation(s)
- Yuman Dong
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, China
- Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Jieru Li
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Yiwei Dai
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Xinyu Zhang
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Xiangyan Jiang
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Tao Wang
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Bin Zhao
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Wenbo Liu
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Haonan Sun
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Pengcheng Du
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Long Qin
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, China
- Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Zuoyi Jiao
- Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
| |
Collapse
|
72
|
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
|
73
|
Liu S, Chen J, Li L, Ye Z, Liu J, Chen Y, Hu B, Tang J, Feng G, Li Z, Deng C, Deng R, Zhu X, Zhang H. Susceptibility of Mitophagy-Deficient Tumors to Ferroptosis Induction by Relieving the Suppression of Lipid Peroxidation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412593. [PMID: 39679775 PMCID: PMC11809388 DOI: 10.1002/advs.202412593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/27/2024] [Indexed: 12/17/2024]
Abstract
The identification of ferroptosis-sensitive cancers is critical for the application of ferroptosis-inducing therapies in cancer therapy. Here, patient-derived organoid screening models of colorectal cancer are established to identify tumors that are sensitive to ferroptosis-inducing therapy. This study discovers that patient-derived tumors characterized by mitophagy deficiency are hypersensitive to ferroptosis-inducing therapies. Mechanistically, a novel negative feedback regulatory pathway of lipid peroxidation is identified, which is one of the important intrinsic anti-ferroptosis mechanisms of cancer cells. Lipid peroxidation-mediated endoplasmic reticulum stress transcriptionally upregulates Parkin to promote mitophagy through ATF4. Mitophagy limits the generation of lipid peroxidation products and subsequently inhibits ferroptosis by inhibiting the accumulation of mitochondrial ROS. Mitophagy-deficient tumors lack this anti-ferroptotic mechanism, unleashing the generation of lipid peroxidation and potent ferroptotic cell death induced by erastin, RSL3, cysteine deprivation, radiotherapy, and immunotherapy. More importantly, ferroptosis-inducing therapy selectively inhibits the growth and distant metastasis of mitophagy-deficient tumors in vivo. In summary, patient-derived organoids of colorectal cancer patients for screening ferroptosis-sensitive tumors are established, providing a paradigm for identifying that patient-derived tumors are sensitive to ferroptosis-inducing therapies. This study concludes that mitophagy-deficient tumors are vulnerable to ferroptosis induction, which may lead to the development of new therapeutic strategies for tumors deficient in mitophagy.
Collapse
Affiliation(s)
- Shan Liu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
- Department of Medical OncologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhen518107China
- Department of Medical OncologySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Jing‐Hong Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and DegradationSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
- Faculty of Health SciencesUniversity of MacauMacau SAR999078China
| | - Li‐Chao Li
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Zhi‐Peng Ye
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Jian‐Nan Liu
- Department of OncologyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantai264000China
| | - Yu‐Hong Chen
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Bing‐Xin Hu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Jia‐Hong Tang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Gong‐Kan Feng
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Zhi‐Ming Li
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
- Department of Medical OncologySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Chu‐Xia Deng
- Faculty of Health SciencesUniversity of MacauMacau SAR999078China
| | - Rong Deng
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Xiao‐Feng Zhu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Hai‐Liang Zhang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerGuangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| |
Collapse
|
74
|
Kai J, Liu X, Wu M, Liu P, Lin M, Yang H, Zhao Q. Technological advances in clinical individualized medication for cancer therapy: from genes to whole organism. Per Med 2025; 22:45-58. [PMID: 39764674 DOI: 10.1080/17410541.2024.2447224] [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: 02/28/2024] [Accepted: 12/23/2024] [Indexed: 02/13/2025]
Abstract
Efforts have been made to leverage technology to accurately identify tumor characteristics and predict how each cancer patient may respond to medications. This involves collecting data from various sources such as genomic data, histological information, functional drug profiling, and drug metabolism using techniques like polymerase chain reaction, sanger sequencing, next-generation sequencing, fluorescence in situ hybridization, immunohistochemistry staining, patient-derived tumor xenograft models, patient-derived organoid models, and therapeutic drug monitoring. The utilization of diverse detection technologies in clinical practice has made "individualized treatment" possible, but the desired level of accuracy has not been fully attained yet. Here, we briefly summarize the conventional and state-of-the-art technologies contributing to individualized medication in clinical settings, aiming to explore therapy options enhancing clinical outcomes.
Collapse
Affiliation(s)
- Jiejing Kai
- Department of Clinical Pharmacy, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xueling Liu
- Department of Clinical Pharmacy, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meijia Wu
- Department of Clinical Pharmacy, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pan Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Meihua Lin
- Department of Clinical Pharmacy, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongyu Yang
- Department of Clinical Pharmacy, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingwei Zhao
- Department of Clinical Pharmacy, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
75
|
Zhang W, Wu C, Huang H, Bleu P, Zambare W, Alvarez J, Wang L, Paty PB, Romesser PB, Smith JJ, Chen XS. Enhancing chemotherapy response prediction via matched colorectal tumor-organoid gene expression analysis and network-based biomarker selection. Transl Oncol 2025; 52:102238. [PMID: 39754813 PMCID: PMC11754497 DOI: 10.1016/j.tranon.2024.102238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/25/2024] [Accepted: 12/07/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) presents significant challenges in chemotherapy response prediction due to its molecular heterogeneity. Current methods often fail to account for the complexity and variability inherent in individual tumors. METHODS We developed a novel approach using matched CRC tumor and organoid gene expression data. We applied Consensus Weighted Gene Co-expression Network Analysis (WGCNA) across three datasets: CRC tumors, matched organoids, and an independent organoid dataset with IC50 drug response values, to identify key gene modules and hub genes linked to chemotherapy response, particularly 5-fluorouracil (5-FU). FINDINGS Our integrative analysis identified significant gene modules and hub genes associated with CRC chemotherapy response. The predictive model built from these findings demonstrated superior accuracy over traditional methods when tested on independent datasets. The matched tumor-organoid data approach proved effective in capturing relevant biomarkers, enhancing prediction reliability. INTERPRETATION This study provides a robust framework for improving CRC chemotherapy response predictions by leveraging matched tumor and organoid gene expression data. Our approach addresses the limitations of previous methods, offering a promising strategy for personalized treatment planning in CRC. Future research should aim to validate these findings and explore the integration of more comprehensive drug response data. FUNDING This research was supported by US National Cancer Institute grant R37CA248289, and Sylvester Comprehensive Cancer Center. which receives funding from the National Cancer Institute award P30CA240139. This work was supported by National Institutes of Health (NIH) under the following grants: T32CA009501-31A1 and R37CA248289. This work was also supported by the MSK P30CA008748 grant.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Chao Wu
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hanchen Huang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Paulina Bleu
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wini Zambare
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Janet Alvarez
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lily Wang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Philip B Paty
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Paul B Romesser
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - J Joshua Smith
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - X Steven Chen
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
76
|
Aksoy SA, Earl J, Grahovac J, Karakas D, Lencioni G, Sığırlı S, Bijlsma MF. Organoids, tissue slices and organotypic cultures: Advancing our understanding of pancreatic ductal adenocarcinoma through in vitro and ex vivo models. Semin Cancer Biol 2025; 109:10-24. [PMID: 39730107 DOI: 10.1016/j.semcancer.2024.12.003] [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: 10/04/2024] [Revised: 12/14/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has one of the worst prognoses of all common solid cancers. For the large majority of PDAC patients, only systemic therapies with very limited efficacy are indicated. In addition, immunotherapies have not brought the advances seen in other cancer types. Several key characteristics of PDAC contribute to poor treatment outcomes, and in this review, we will discuss how these characteristics are best captured in currently available ex vivo or in vitro model systems. For instance, PDAC is hallmarked by a highly desmoplastic and immune-suppressed tumor microenvironment that impacts disease progression and therapy resistance. Also, large differences in tumor biology exist between and within tumors, complicating treatment decisions. Furthermore, PDAC has a very high propensity for locally invasive and metastatic growth. The use of animal models is often not desirable or feasible and several in vitro and ex vivo model systems have been developed, such as organotypic cocultures and tissue slices, among others. However, the absence of a full host organism impacts the ability of these models to accurately capture the characteristics that contribute to poor outcomes in PDAC. We will discuss the caveats and advantages of these model systems in the context of PDAC's key characteristics and provide recommendations on model choice and the possibilities for optimization. These considerations should be of use to researchers aiming to study PDAC in the in vitro setting.
Collapse
Affiliation(s)
- Secil Ak Aksoy
- Bursa Uludag University, Faculty of Medicine, Department of Medical Microbiology, Bursa, Turkey
| | - Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Biomodels and Biomodels Platform Hospital Ramón y Cajal-IRYCIS, Carretera Colmenar Km 9,100, Madrid 28034, Spain; The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid 28029, Spain
| | - Jelena Grahovac
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Didem Karakas
- Acibadem Mehmet Ali Aydinlar University, Department of Medical Biotechnology, Graduate School of Health Sciences, Istanbul, Turkey
| | - Giulia Lencioni
- Department of Biology, University of Pisa, Pisa, Italy; Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
| | - Sıla Sığırlı
- Acibadem Mehmet Ali Aydinlar University, Department of Medical Biotechnology, Graduate School of Health Sciences, Istanbul, Turkey
| | - Maarten F Bijlsma
- Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands.
| |
Collapse
|
77
|
Wang Y, Zhang Z, Li X, Lu X, Zhuansun X, Li Q, Zhang J, Xu X, Liu X, Wei Y, Hua F, Wu R, Chen Z. Colorectal carcinoma organoid and cancer-associated fibroblasts co-culture system for drug evaluation. IN VITRO MODELS 2025; 4:31-44. [PMID: 40160212 PMCID: PMC11950461 DOI: 10.1007/s44164-025-00084-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 04/02/2025]
Abstract
Patient-derived organoids (PDO) have the potential to be used as preclinical cancer models for testing anti-cancer drug efficiency. Cancer-associated fibroblasts (CAFs), which have been closely linked with colorectal carcinoma (CRC) progression and drug resistance, however, are generally not included (or gradually lost during culture) in the PDO models, leading to a major limitation in this cancer model. In this study, we established a new in vitro model with CRC organoids and co-cultured with CAFs and compared it with the organoid-only model. Through testing with anti-cancer drug, we demonstrated a significant difference in drug sensitivity between the two models, and the co-culture model showed higher drug resistance. RNA and whole exome sequencing were performed to reveal gene expression profiles in organoids and organoids co-culture with CAFs to assess interactions between drug sensitivity and gene copy number variation. We found that the expression levels of several pathway protein genes, which are highly expressed in original surgical specimens of colorectal carcinomas, were downregulated in organoids but restored in organoids by co-culturing with CAFs. In summary, the PDO-CAF joint model for CRC can recapitulate a more biomimetic tumor microenvironment and the drug resistance lead by changes in multiple signaling pathways that we discovered; thus, it could be a suitable model for future usage in drug discovery and precision medicine research.
Collapse
Affiliation(s)
- Yan Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, 215163 China
| | - Zilin Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Xiaoran Li
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, 215163 China
- Jiangsu Avatarget Biotechnology Co., Ltd, Suzhou, 215163 China
| | - Xiaobing Lu
- Jiangsu Health Vocational College, Nanjing, 210029 China
| | - Xuemei Zhuansun
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, 215163 China
- Jiangsu Avatarget Biotechnology Co., Ltd, Suzhou, 215163 China
| | - Qiwei Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Jing Zhang
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, 215163 China
- Jiangsu Avatarget Biotechnology Co., Ltd, Suzhou, 215163 China
| | - Xi Xu
- Jiangsu Avatarget Biotechnology Co., Ltd, Suzhou, 215163 China
| | - Xueqiang Liu
- Jiangsu Avatarget Biotechnology Co., Ltd, Suzhou, 215163 China
| | - Yuan Wei
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, 215163 China
- Jiangsu Avatarget Biotechnology Co., Ltd, Suzhou, 215163 China
| | - Feng Hua
- Department of Pharmacy, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, 213000 China
| | - Runda Wu
- The First Affiliated Hospital of Suzhou University, Suzhou, 215006 China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, 215163 China
| |
Collapse
|
78
|
McGreevy O, Bosakhar M, Gilbert T, Quinn M, Fenwick S, Malik H, Goldring C, Randle L. The importance of preclinical models in cholangiocarcinoma. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:108304. [PMID: 38653585 DOI: 10.1016/j.ejso.2024.108304] [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: 03/20/2024] [Accepted: 03/23/2024] [Indexed: 04/25/2024]
Abstract
Cholangiocarcinoma (CCA) is an adenocarcinoma of the hepatobiliary system with a grim prognosis. Incidence is rising globally and surgery is currently the only curative treatment, but is only available for patients who are fit and diagnosed in an early-stage of disease progression. Great importance has been placed on developing preclinical models to help further our understanding of CCA and potential treatments to improve therapeutic outcomes. Preclinical models of varying complexity and cost have been established, ranging from more simplistic in vitro 2D CCA cell lines in culture, to more complex in vivo genetically engineered mouse models. Currently there is no single model that faithfully recaptures the complexities of human CCA and the in vivo tumour microenvironment. Instead a multi-model approach should be used when designing preclinical trials to study CCA and potential therapies.
Collapse
Affiliation(s)
- Owen McGreevy
- The Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Mohammed Bosakhar
- The Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Timothy Gilbert
- The Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK; Hepatobiliary Surgery, Liverpool University Hospitals NHS Foundation Trust, Royal Liverpool University Hospital, Prescot Street, L7 8XP, Liverpool, UK
| | - Marc Quinn
- The Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK; Hepatobiliary Surgery, Liverpool University Hospitals NHS Foundation Trust, Royal Liverpool University Hospital, Prescot Street, L7 8XP, Liverpool, UK
| | - Stephen Fenwick
- Hepatobiliary Surgery, Liverpool University Hospitals NHS Foundation Trust, Royal Liverpool University Hospital, Prescot Street, L7 8XP, Liverpool, UK
| | - Hassan Malik
- Hepatobiliary Surgery, Liverpool University Hospitals NHS Foundation Trust, Royal Liverpool University Hospital, Prescot Street, L7 8XP, Liverpool, UK
| | - Christopher Goldring
- The Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Laura Randle
- The Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK.
| |
Collapse
|
79
|
Paul CD, Yankaskas C, Shahi Thakuri P, Balhouse B, Salen S, Bullock A, Beam S, Chatman A, Djikeng S, Yang XJ, Wong G, Dey I, Holmes S, Dockey A, Bailey-Steinitz L, Zheng L, Li W, Chandra V, Nguyen J, Sharp J, Willems E, Kennedy M, Dallas MR, Kuninger D. Long-term maintenance of patient-specific characteristics in tumoroids from six cancer indications. Sci Rep 2025; 15:3933. [PMID: 39890889 PMCID: PMC11785764 DOI: 10.1038/s41598-025-86979-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025] Open
Abstract
Tumoroids, sometimes referred to as cancer organoids, are patient-derived cancer cells grown as 3D, self-organized multicellular structures that maintain key characteristics (e.g., genotype, gene expression levels) of the tumor from which they originated. These models have emerged as valuable tools for studying tumor biology, cytotoxicity, and response of patient-derived cells to cancer therapies. However, the establishment and maintenance of tumoroids has historically been challenging, labor intensive, and highly variable from lab to lab, hindering their widespread use. Here, we characterize the establishment and/or expansion of colorectal, lung, head and neck, breast, pancreas, and endometrial tumoroids using the standardized, serum-free Gibco OncoPro Tumoroid Culture Medium. Newly derived tumoroid lines (n = 20) were analyzed by targeted genomic profiling and RNA sequencing and were representative of tumor tissue samples. Tumoroid lines were stable for over 250 days in culture and freeze-thaw competent. Previously established tumoroid lines were also transitioned to OncoPro medium and exhibited, on average, similar growth rates and conserved donor-specific characteristics when compared to original media systems. Additionally, OncoPro medium was compatible with both embedded culture in extracellular matrix and growth in a suspension format for facile culture and scale up. An example application of these models for assessing the cytotoxicity of a natural killer cell line and primary natural killer cells over time and at various doses demonstrated the compatibility of these models with assays used in compound and cell therapy development. We anticipate that the standardization and versatility of this approach will have important benefits for basic cancer research, drug discovery, and personalized medicine and help make tumoroid models more accessible to the cancer research community.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sylvia Beam
- Thermo Fisher Scientific, Frederick, MD, USA
| | | | | | | | | | - Isha Dey
- Thermo Fisher Scientific, Bengaluru, Karnataka, India
| | | | | | | | - Lina Zheng
- Thermo Fisher Scientific, Carlsbad, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Palacios PA, Flores I, Cereceda L, Otero FF, Müller M, Brebi P, Contreras HR, Carreño LJ. Patient-Derived Organoid Models for NKT Cell-Based Cancer Immunotherapy. Cancers (Basel) 2025; 17:406. [PMID: 39941775 PMCID: PMC11815936 DOI: 10.3390/cancers17030406] [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: 12/12/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Invariant Natural Killer T (iNKT) cells are a unique subset of T cells that bridge innate and adaptive immunity, displaying potent anti-tumor properties through cytokine secretion, direct cytotoxicity, and recruitment of immune effector cells such as CD8+ T cells and NK cells. Despite their therapeutic potential, the immunosuppressive tumor microenvironment (TME), characterized by regulatory T cells, myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs), limits iNKT cell efficacy. Patient-derived organoid (PDO) platforms provide an innovative model for dissecting these complex interactions and evaluating strategies to reinvigorate iNKT cell functionality within the TME. PDOs closely mimic the genetic, phenotypic, and structural characteristics of primary tumors, enabling the study of tumor-immune dynamics. Integrating iNKT cells into PDOs offers a robust platform for investigating CD1d-mediated interactions, Th1-biased immune responses driven by glycolipid analogs like α-GalCer, and combination therapies such as immune checkpoint inhibitors. Additionally, PDO systems can assess the effects of metabolic modulation, including reducing lactic acid accumulation or targeting glutamine pathways, on enhancing iNKT cell activity. Emerging innovations, such as organoid-on-a-chip systems, CRISPR-Cas9 gene editing, and multi-omics approaches, further expand the potential of PDO-iNKT platforms for personalized immunotherapy research. Although the application of iNKT cells in PDOs is still undeveloped, these systems hold immense promise for bridging preclinical studies and clinical translation. By addressing the challenges of the TME and optimizing therapeutic strategies, PDO-iNKT platforms offer a transformative avenue for advancing cancer immunotherapy and personalized medicine.
Collapse
Affiliation(s)
- Pablo A. Palacios
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Iván Flores
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8350499, Chile
| | - Lucas Cereceda
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Francisco F. Otero
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Marioly Müller
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Priscilla Brebi
- Millennium Institute on Immunology and Immunotherapy, Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
- Biomedical Research Consortium (BMRC), Santiago 8331150, Chile
| | - Héctor R. Contreras
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8350499, Chile
- Center for Cancer Prevention and Control (CECAN), Santiago 8350499, Chile
| | - Leandro J. Carreño
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Biomedical Research Consortium (BMRC), Santiago 8331150, Chile
| |
Collapse
|
81
|
Zhang Y, Jin Z, Wang Z, Yan L, Liu A, Li F, Li Y, Zhang Y. Trends in Colorectal Cancer Peritoneal Metastases Research: A Comprehensive Bibliometric Analysis. J Gastrointest Cancer 2025; 56:51. [PMID: 39847239 DOI: 10.1007/s12029-025-01176-1] [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] [Accepted: 01/11/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) stands as the third most prevalent malignancy globally and is recognized as the second leading cause of cancer-related mortality. Notably, nearly 50% of individuals diagnosed with CRC ultimately develop metastatic disease, with the peritoneum emerging as the second most frequent site for metastatic spread. Recent advancements in therapeutic frameworks have enhanced both survival rates and quality of life metrics for patients afflicted with colorectal cancer peritoneal metastases (CRCPM). OBJECTIVE This study endeavors to facilitate an in-depth review of the current scientific landscape surrounding CRCPM, ultimately aiming to delineate future avenues for investigative research in this realm. METHODS Employing R software through the Bibliometrix package, alongside analytical tools such as CiteSpace and VOSviewer, we performed a comprehensive bibliometric analysis. This enabled us to assess pivotal keywords, prominent authors, influential countries, notable institutions, relevant literature, and key journals pertinent to the field of CRCPM research. RESULTS Our findings illustrate a significant uptick in the volume of publications addressing CRCPM, with the USA leading in overall contribution, complemented by substantial input from distinguished scholars in the Netherlands and France. The author Ignace H. J. T. de Hingh emerged as the most prolific contributor. Current research endeavors have predominantly focused on the characterization of primary malignancies with peritoneal metastases, therapeutic interventions for CRCPM, and the orchestration of clinical trials. CONCLUSION This analysis culminates in a systematic encapsulation of the prevailing research findings concerning CRCPM, underscoring current hotspots and predicting future trends within the global research spectrum. The exploration of treatment modalities for CRCPM remains vibrant, and ongoing multicenter clinical trials are anticipated to further enrich our understanding and management of this challenging clinical issue.
Collapse
Affiliation(s)
- Yuzhe Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Zi Jin
- Nuclear Medicine Department, Shenyang Fifth People's Hospital, Shenyang, 110001, China
| | - Zhongqing Wang
- Department of Information Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Lirong Yan
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Aoran Liu
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Fang Li
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yanke Li
- Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang, 110001, China.
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
82
|
Grigoreva TA, Kindt DN, Sagaidak AV, Novikova DS, Tribulovich VG. Cellular Systems for Colorectal Stem Cancer Cell Research. Cells 2025; 14:170. [PMID: 39936962 PMCID: PMC11817814 DOI: 10.3390/cells14030170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Oncological diseases consistently occupy leading positions among the most life-threatening diseases, including in highly developed countries. At the same time, the second most common cause of cancer death is colorectal cancer. The current level of research shows that the development of effective therapy, in this case, requires a new grade of understanding processes during the emergence and development of a tumor. In particular, the concept of cancer stem cells that ensure the survival of chemoresistant cells capable of giving rise to new tumors is becoming widespread. To provide adequate conditions that reproduce natural processes typical for tumor development, approaches based on increasingly complex cellular systems are being improved. This review discusses the main strategies that allow for the study of the properties of tumor cells with an emphasis on colorectal cancer stem cells. The features of working with tumor cells and the advantages and disadvantages of 2D and 3D culture systems are considered.
Collapse
Affiliation(s)
- Tatyana A. Grigoreva
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia (V.G.T.)
| | | | | | | | | |
Collapse
|
83
|
Sui Z, Wu X, Wang J, Tan S, Zhao C, Yu Z, Wu C, Wang X, Guo L. Mesenchymal stromal cells promote the formation of lung cancer organoids via Kindlin-2. Stem Cell Res Ther 2025; 16:7. [PMID: 39789648 PMCID: PMC11715222 DOI: 10.1186/s13287-024-04128-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Patient-derived lung cancer organoids (PD-LCOs) demonstrate exceptional potential in preclinical testing and serve as a promising model for the multimodal management of lung cancer. However, certain lung cancer cells derived from patients exhibit limited capacity to generate organoids due to inter-tumor or intra-tumor variability. To overcome this limitation, we have created an in vitro system that employs mesenchymal stromal cells (MSCs) or fibroblasts to serve as a supportive scaffold for lung cancer cells that do not form organoids. METHODS We successfully established an MSCs/fibroblast co-culture system to form LCOs. We analyzed the morphological and histological similarities between LCOs co-cultured with fibroblast and primary lung cancer lesions through HE and IF staining. We evaluated whether LCOs co-cultured with fibroblast retained the original genetic mutations of their source tumors based on WES. RNA sequencing was used to analyze the differences in gene expression profiles between LCOs co-cultured with fibroblast and paracancerous organoids (POs). Importantly, we have successfully validated the impact of Kindlin-2 on the regulation of MSCs in organoid formation through lentiviral vector-mediated interference or overexpression of kindlin-2. RESULTS Our findings demonstrate that the addition of MSCs/fibroblasts to three tumor samples, initially incapable of forming organoids by traditional methods, successfully facilitated the cultivation of tumor organoids. Importantly, these organoids co-cultured with fibroblast faithfully recapitulate the tissue morphology of original lung tumors and replicate the genetic profile observed in the parental tumors even after prolonged in vitro culture. Moreover, drug responses exhibited by these organoids co-cultured with MSCs/fibroblasts are consistent with those observed in the original tumors. Mechanistically, we have also identified kindlin-2 as a crucial regulator linking extracellular matrix (ECM) and mitochondria that influence MSC/fibroblast-mediated support for tumor organoid formation. CONCLUSION The results obtained from our research enhance the understanding of the mechanisms implicated in the formation of tumor organoids and aid in creating stronger patient-specific tumor organoid models. This advancement supports the refinement of personalized drug response assessments for use in clinical settings.
Collapse
Affiliation(s)
- Zhilin Sui
- Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Xianxian Wu
- Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Jiaxin Wang
- Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - ShihJye Tan
- Department of Biology, and Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chao Zhao
- Institute of Scientific Instrumentation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zhentao Yu
- Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Chuanyue Wu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
| | - Xiaoxiao Wang
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China.
- Department of Biology, and Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Ling Guo
- Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China.
- Department of Biology, and Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
84
|
Zhang Y, Lian X, Xu H, Zhu S, Zhang H, Ni Z, Fu T, Liu S, Tao L, Zhou Y, Zhu F. OrgXenomics: an integrated proteomic knowledge base for patient-derived organoid and xenograft. Nucleic Acids Res 2025; 53:D504-D515. [PMID: 39373514 PMCID: PMC11701540 DOI: 10.1093/nar/gkae861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024] Open
Abstract
Patient-derived models (PDMs, particularly organoids and xenografts) are irreplaceable tools for precision medicine, from target development to lead identification, then to preclinical evaluation, and finally to clinical decision-making. So far, PDM-based proteomics has emerged to be one of the cutting-edge directions and massive data have been accumulated. However, such PDM-based proteomic data have not been provided by any of the available databases, and proteomics profiles of all proteins in proteomic study are also completely absent from existing databases. Herein, an integrated database named 'OrgXenomics' was thus developed to provide the proteomic data for PDMs, which was unique in (a) explicitly describing the establishment detail for a wide array of models, (b) systematically providing the proteomic profiles (expression/function/interaction) for all proteins in studied proteomic analysis and (c) comprehensively giving the raw data for diverse organoid/xenograft-based proteomic studies of various diseases. Our OrgXenomics was expected to server as one good complement to existing proteomic databases, and had great implication for the practice of precision medicine, which could be accessed at: https://idrblab.org/orgxenomics/.
Collapse
Affiliation(s)
- Yintao Zhang
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Xichen Lian
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Hangwei Xu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Sisi Zhu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Hao Zhang
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Ziheng Ni
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Tingting Fu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Shuiping Liu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Lin Tao
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
85
|
Chen J, Cheng S, Gu L, Huang Z, Zhang C, Sun C, Chen S. Establishment and characterization of a sigmoid colon cancer organoid with spinal metastasis. Front Cell Dev Biol 2025; 12:1510264. [PMID: 39830210 PMCID: PMC11739105 DOI: 10.3389/fcell.2024.1510264] [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: 10/12/2024] [Accepted: 11/29/2024] [Indexed: 01/22/2025] Open
Abstract
Background Sigmoid colon cancer with spinal metastases is rare in distant metastasis. In addition, the prognosis of colon cancer patients with spinal metastases is extremely poor. In order to find effective therapeutic agents, we need to know the biological characteristics of such patients from related models. Methods We collected sigmoid colon cancer tissue from a young female subject who was diagnosed with sigmoid colon cancer with multiple spinal metastases. We successfully established a sigmoid colon cancer organoid using this tissue and investigated drug screening in the patient. HE staining, immunohistochemistry, and DNA sequencing were utilized to compare the biological characteristics between the original tumor and the organoid. Furthermore, we investigated the drug screening of the sigmoid colon cancer organoid in vitro. Results A colon cancer organoid from sigmoid colon cancer with spinal metastases was successfully established. The organoid culture maintained the morphological features, histological features, and genomic landscape of the corresponding sigmoid colon cancer cells. Moreover, we performed drug screening tests to evaluate the effects of chemotherapeutic drugs and targeted drugs. Conclusion The sigmoid colon cancer organoid with spinal metastases was a favorable preclinical model to explore the clinicopathologic characteristics of colon cancer patients with spinal metastases.
Collapse
Affiliation(s)
- Jingyao Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shumin Cheng
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Liang Gu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhangsen Huang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chunhui Sun
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Songyao Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
86
|
Anandi L, Garcia J, Ros M, Janská L, Liu J, Carmona-Fontaine C. Direct visualization of emergent metastatic features within an ex vivo model of the tumor microenvironment. Life Sci Alliance 2025; 8:e202403053. [PMID: 39419548 PMCID: PMC11487089 DOI: 10.26508/lsa.202403053] [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: 09/19/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Ischemic conditions such as hypoxia and nutrient starvation, together with interactions with stromal cells, are critical drivers of metastasis. These conditions arise deep within tumor tissues, and thus, observing nascent metastases is exceedingly challenging. We thus developed the 3MIC-an ex vivo model of the tumor microenvironment-to study the emergence of metastatic features in tumor cells in a 3-dimensional (3D) context. Here, tumor cells spontaneously create ischemic-like conditions, allowing us to study how tumor spheroids migrate, invade, and interact with stromal cells under different metabolic conditions. Consistent with previous data, we show that ischemia increases cell migration and invasion, but the 3MIC allowed us to directly observe and perturb cells while they acquire these pro-metastatic features. Interestingly, our results indicate that medium acidification is one of the strongest pro-metastatic cues and also illustrate using the 3MIC to test anti-metastatic drugs on cells experiencing different metabolic conditions. Overall, the 3MIC can help dissecting the complexity of the tumor microenvironment for the direct observation and perturbation of tumor cells during the early metastatic process.
Collapse
Affiliation(s)
- Libi Anandi
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Jeremy Garcia
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Manon Ros
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Libuše Janská
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Josephine Liu
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Carlos Carmona-Fontaine
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| |
Collapse
|
87
|
Abbasian MH, Sobhani N, Sisakht MM, D'Angelo A, Sirico M, Roudi R. Patient-Derived Organoids: A Game-Changer in Personalized Cancer Medicine. Stem Cell Rev Rep 2025; 21:211-225. [PMID: 39432173 DOI: 10.1007/s12015-024-10805-4] [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] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Research on cancer therapies has benefited from predictive tools capable of simulating treatment response and other disease characteristics in a personalized manner, in particular three-dimensional cell culture models. Such models include tumor-derived spheroids, multicellular spheroids including organotypic multicellular spheroids, and tumor-derived organoids. Additionally, organoids can be grown from various cancer cell types, such as pluripotent stem cells and induced pluripotent stem cells, progenitor cells, and adult stem cells. Although patient-derived xenografts and genetically engineered mouse models replicate human disease in vivo, organoids are less expensive, less labor intensive, and less time-consuming, all-important aspects in high-throughput settings. Like in vivo models, organoids mimic the three-dimensional structure, cellular heterogeneity, and functions of primary tissues, with the advantage of representing the normal oxygen conditions of patient organs. In this review, we summarize the use of organoids in disease modeling, drug discovery, toxicity testing, and precision oncology. We also summarize the current clinical trials using organoids.
Collapse
Affiliation(s)
- Mohammad Hadi Abbasian
- Department of Medical Genetics, National Institute for Genetic Engineering and Biotechnology, Tehran, Iran
| | - Navid Sobhani
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Mahsa Mollapour Sisakht
- Faculty of Pharmacy, Biotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Alberto D'Angelo
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AX, UK
| | - Marianna Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Raheleh Roudi
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
88
|
Li Y, Wu W, Yao J, Wang S, Wu X, Yan J. Patient-Derived Tumor Organoids: A Platform for Precision Therapy of Colorectal Cancer. Cell Transplant 2025; 34:9636897251314645. [PMID: 39953837 PMCID: PMC11829288 DOI: 10.1177/09636897251314645] [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: 10/31/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 02/17/2025] Open
Abstract
Colorectal cancer (CRC) represents a significant cause of cancer-related mortality on a global scale. It is a highly heterogeneous cancer, and the response of patients to homogeneous drug therapy varies considerably. Patient-derived tumor organoids (PDTOs) represent an optimal preclinical model for cancer research. A substantial body of evidence from numerous studies has demonstrated that PDTOs can accurately predict a patient's response to different drug treatments. This article outlines the utilization of PDTOs in the management of CRC across a range of therapeutic contexts, including postoperative adjuvant chemotherapy, palliative chemotherapy, neoadjuvant chemoradiotherapy, targeted therapy, third-line and follow-up treatment, and the treatment of elderly patients. This article delineates the manner in which PDTOs can inform therapeutic decisions at all stages of CRC, thereby assisting clinicians in selecting treatment options and reducing the risk of toxicity and resistance associated with clinical drugs. Moreover, it identifies shortcomings of existing PDTOs, including the absence of consistent criteria for assessing drug sensitivity tests, the lack of vascular and tumor microenvironment models, and the high cost of the technology. In conclusion, despite their inherent limitations, PDTOs offer several advantages, including rapid culture, a high success rate, high consistency, and high throughput, which can be employed as a personalized treatment option for CRC. The use of PDTOs in CRC allows for the prediction of responses to different treatment modalities at various stages of disease progression. This has the potential to reduce adverse drug reactions and the emergence of resistance associated with clinical drugs, facilitate evidence-based clinical decision-making, and guide CRC patients in the selection of personalized medications, thereby advancing the individualized treatment of CRC.
Collapse
Affiliation(s)
- Yiran Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Wei Wu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Jiaxin Yao
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Suidong Wang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Xiufeng Wu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, P.R. China
| | - Jun Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| |
Collapse
|
89
|
Zhang Q, He J, Zhu D, Chen Y, Fu M, Lu S, Qiu Y, Zhou G, Yang G, Jiang Z. Genetically modified organoids for tissue engineering and regenerative medicine. Adv Colloid Interface Sci 2025; 335:103337. [PMID: 39547125 DOI: 10.1016/j.cis.2024.103337] [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: 01/18/2024] [Revised: 07/23/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
To date, genetically modified organoids are emerging as a promising 3D modeling tool aimed at solving genetically relevant clinical and biomedical problems for regenerative medicine and tissue engineering. As an optimal vehicle for gene delivery, genetically modified organoids can enhance or reduce the expression of target genes through virus and non-virus-based gene transfection methods to achieve tissue regeneration. Animal experiments and preclinical studies have demonstrated the beneficial role of genetically modified organoids in various aspects of organ regeneration, including thymus, lacrimal glands, brain, lung, kidney, photoreceptors, etc. Furthermore, the technology offers a potential treatment option for various diseases, such as Fabry disease, non-alcoholic steatohepatitis, and Lynch syndrome. Nevertheless, the uncertain safety of genetic modification, the risk of organoid application, and bionics of current genetically modified organoids are still challenging. This review summarizes the researches on genetically modified organoids in recent years, and describes the transfection methods and functions of genetically modified organoids, then introduced their applications at length. Also, the limitations and future development directions of genetically modified organoids are included.
Collapse
Affiliation(s)
- Qinmeng Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Jin He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Danji Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Yunxuan Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Mengdie Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Shifan Lu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Yuesheng Qiu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Guodong Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
90
|
Wang B, Hasturk O, Kumarasinghe U, Rudolph S, Staii C, Chen Y, Kaplan DL. Temporary Nanoencapsulation of Human Intestinal Organoids Using Silk Ionomers. Adv Healthc Mater 2025; 14:e2403176. [PMID: 39648539 DOI: 10.1002/adhm.202403176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/03/2024] [Indexed: 12/10/2024]
Abstract
Human intestinal organoids (HIOs) are vital for modeling intestinal development, disease, and therapeutic tissue regeneration. However, their susceptibility to stress, immunological attack, and environmental fluctuations limits their utility in research and therapeutic applications. This study evaluated the effectiveness of temporary silk protein-based layer-by-layer (LbL) nanoencapsulation technique to enhance the viability and functions of HIOs against common biomedical stressors, without compromising their native functions. Cell viability and differentiation capacity are assessed, finding that nanoencapsulation significantly improved HIO survival under the various environmental perturbations studied without compromising cellular functionality. Post-stress exposures, the encapsulated HIOs still successfully differentiated into essential intestinal cell types such as enterocytes, goblet cells, enteroendocrine cells, and Paneth cells. Moreover, the silk nanocoatings effectively protected against environmental stressors such as ultraviolet (UV) light exposure, protease degradation, antibody binding, and cytokine-induced inflammation. This nanoencapsulation technique shows promise for advancing HIO applications in disease modeling, drug testing, and potential transplantation therapies.
Collapse
Affiliation(s)
- Brooke Wang
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Onur Hasturk
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | | | - Sara Rudolph
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Cristian Staii
- Department of Physics and Astronomy, Tufts University, Medford, MA, USA
| | - Ying Chen
- Department of Physics and Astronomy, Tufts University, Medford, MA, USA
| | - David L Kaplan
- Department of Physics and Astronomy, Tufts University, Medford, MA, USA
| |
Collapse
|
91
|
Olvera-Valencia M, Garcia-Castillo V, Ramos-Payan R, Aguilar-Medina M, Trujano-Camacho S, López-Saavedra A, Marchat LA, López-Camarillo C, Sumagin R, Pérez-Yepez E, Pérez-Plasencia C. Development of a reliable method for human triple-negative breast cancer organotypic culture: Improving imaging and genomic studies in 3D cultures. J Tissue Eng 2025; 16:20417314251326668. [PMID: 40342587 PMCID: PMC12059422 DOI: 10.1177/20417314251326668] [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: 11/08/2024] [Accepted: 02/25/2025] [Indexed: 05/11/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive and lacks targeted therapies, posing a major challenge in oncology. Traditional two-dimensional (2D) cell cultures fail to capture the tumor microenvironment's complexity, whereas three-dimensional (3D) cultures provide a more accurate model of tumor biology. We developed an advanced 3D culture system for TNBC cell lines BT-20 and MDA-MB-231, enhancing the hanging-drop method with Matrigel to restore essential extracellular matrix interactions. Confocal imaging showed MDA-MB-231 cells forming clusters typical of aggressive carcinoma, while BT-20 cells organized into duct-like structures. Molecular analysis of PI3K and β-catenin target genes revealed distinct expression patterns, with PI3K overexpressed and β-catenin downregulated in 3D cultures. Moreover, β-catenin distribution in the 3D cell culture closely resembles its pattern in tissue. These findings underscore the value of 3D models in understanding TNBC progression and in supporting the exploration of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mercedes Olvera-Valencia
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía del Instituto Politécnico Nacional, Ticoman, CDMX, Mexico
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX, Mexico
| | - Verónica Garcia-Castillo
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla Estado de México, Mexico
| | - Rosalío Ramos-Payan
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacan, Sinaloa, Mexico
| | - Maribel Aguilar-Medina
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacan, Sinaloa, Mexico
| | - Samuel Trujano-Camacho
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX, Mexico
- Experimental Biology PhD Program, DCBS, Universidad Autónoma Metropolitana- Iztapalapa, Iztapalapa, Mexico
| | - Alejandro López-Saavedra
- Advanced Microscopy Applications Unit (ADMIRA)-Instituto Nacional de Cancerología, Tlalpan, CDMX, Mexico
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Laurence A. Marchat
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía del Instituto Politécnico Nacional, Ticoman, CDMX, Mexico
| | - Cesar López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Benito Juarez, CDMX, Mexico
| | - Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Eloy Pérez-Yepez
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX, Mexico
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla Estado de México, Mexico
| |
Collapse
|
92
|
Rathje F, Sykora MM, Aberger F, Krenn PW. High Efficiency Lentiviral Transduction of Colon Organoids Using Reversible 2D/3D Culture Techniques. Methods Mol Biol 2025; 2905:245-254. [PMID: 40163310 DOI: 10.1007/978-1-0716-4418-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Organoids are a promising research tool for studying tissue development and disease in vitro. While organoids are frequently considered a replacement or complementary model for in vivo mouse experiments, exploiting their full potential often requires genetically engineered mice as a source of transgenic stem cells, also because genetic manipulation of organoids is rather inefficient and cumbersome. Here, we describe an alternative and optimized murine colon organoid manipulation protocol that reversibly and temporarily interrupts the 3D organoid structure for short-term 2D monolayer culture. This approach allows highly efficient viral transduction and genetic manipulation of stem cells in a 2D setting, followed by 3D stem cell embedding and restoration of the original organoid architecture. This method greatly improves the efficiency of lentiviral-mediated genetic manipulation of organoids and increases their potential applications in CRISPR/Cas9 and compound screens, immune-competent co-cultures, and disease modeling.
Collapse
Affiliation(s)
- Florian Rathje
- Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology (CTBI), Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Martina M Sykora
- Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology (CTBI), Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Fritz Aberger
- Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology (CTBI), Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Peter W Krenn
- Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology (CTBI), Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Salzburg, Austria.
| |
Collapse
|
93
|
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
|
94
|
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 PMCID: PMC11686971 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
|
95
|
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
|
96
|
Lu H, Liu H, Yan R, Ma W, Liu H, Liu R, Sun Y, Ye L, Gao P, Jia W, Zhang P, Zhang H. Carbonic anhydrase 2 facilitates sorafenib resistance by counteracting MCT4-mediated intracellular pH dysregulation in HCC. Cell Rep 2024; 43:114996. [PMID: 39607826 DOI: 10.1016/j.celrep.2024.114996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/24/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024] Open
Abstract
Sorafenib, the targeted therapy for hepatocellular carcinoma (HCC), has been utilized in clinics for over a decade. However, its effectiveness is severely hindered by acquired drug resistance, the mechanisms of which remain largely elusive. In this study, we identify that carbonic anhydrase 2 (CA2) is a key regulator of sorafenib resistance. Mechanistically, sorafenib treatment decreases intracellular pH (pHi) by suppressing monocarboxylate transporter 4 (MCT4) expression, while high levels of CA2 counteract MCT4-mediated pHi dysregulation upon sorafenib treatment, maintaining pHi homeostasis to facilitate cell survival and sorafenib resistance. Targeting CA2 re-sensitizes resistant HCC cells to sorafenib both in vitro and in vivo. Importantly, analysis of clinical samples shows a strong correlation between CA2 expression levels and the therapeutic efficacy of sorafenib in HCC patients. Our findings highlight the significance of CA2 in facilitating sorafenib resistance and propose targeting CA2 as a potential strategy for overcoming sorafenib resistance in HCC patients.
Collapse
Affiliation(s)
- Hui Lu
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Department of Clinical Laboratory, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Huiya Liu
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Ronghui Yan
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wenhao Ma
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Haiying Liu
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Rui Liu
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yuchen Sun
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Ling Ye
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Ping Gao
- Southern Medical University, Guangzhou, China
| | - Weidong Jia
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Pinggen Zhang
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui, China.
| | - Huafeng Zhang
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
97
|
Drouin Z, Lévesque F, Mouzakitis K, Labrie M. Current preclinical models of brain metastasis. Clin Exp Metastasis 2024; 42:5. [PMID: 39694975 DOI: 10.1007/s10585-024-10318-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/30/2024] [Indexed: 12/20/2024]
Abstract
Brain metastases (BMs) represent the most prevalent intracranial malignancy within the adult. They are identified in up to 20% of patients with solid tumors and this percentage varies between tumor types and age. Due to the selective permeability of the blood-brain barrier, most anticancer drugs can't reach significant concentrations in the brain, representing a major obstacle to the patients' survival. Furthermore, intra- and inter-patient heterogeneity and the unique brain microenvironment add a layer of complexity to the clinical management of BMs. In the perspective of finding new therapeutic approaches and better understanding the molecular mechanisms involved in brain metastasis, the use of appropriate preclinical models is essential. Here, we review current in vivo, in vitro and ex vivo models for the study of brain metastasis while outlining their advantages and limitations.
Collapse
Affiliation(s)
- Zacharie Drouin
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3201 Rue Jean-Mignault, Sherbrooke, QC, J1E 4K8, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
- Institut de Recherche sur le Cancer de l'Université de Sherbrooke (IRCUS), Sherbrooke, QC, Canada
| | - Flavie Lévesque
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3201 Rue Jean-Mignault, Sherbrooke, QC, J1E 4K8, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
- Institut de Recherche sur le Cancer de l'Université de Sherbrooke (IRCUS), Sherbrooke, QC, Canada
| | - Korina Mouzakitis
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3201 Rue Jean-Mignault, Sherbrooke, QC, J1E 4K8, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
- Institut de Recherche sur le Cancer de l'Université de Sherbrooke (IRCUS), Sherbrooke, QC, Canada
| | - Marilyne Labrie
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3201 Rue Jean-Mignault, Sherbrooke, QC, J1E 4K8, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada.
- Institut de Recherche sur le Cancer de l'Université de Sherbrooke (IRCUS), Sherbrooke, QC, Canada.
| |
Collapse
|
98
|
Budeus B, Kroepel C, Stasch LM, Klein D. Matrix-free human lung organoids derived from induced pluripotent stem cells to model lung injury. Stem Cell Res Ther 2024; 15:468. [PMID: 39696649 DOI: 10.1186/s13287-024-04106-3] [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: 09/11/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Organoids, as near-physiological 3D culture systems, offer new opportunities to study the pathogenesis of various organs in mimicking the cellular complexity and functionality of human organs. METHOD Here we used a quite simple and very practicable method to successfully generate induced pluripotent stem cell (iPSC)-derived human lung organoids (LuOrg) in a matrix-free manner as an alternative to the widely used preclinical mouse models in order to investigate normal lung damage in detail and as close as possible to the patient. We performed detailed morphological and molecular analyses, including bulk and single cell RNA sequencing, of generated lung organoids and evaluated the quality and robustness of our model as a potential in vitro platform for lung diseases, namely radiation-induced lung injury. RESULTS A matrix-free method for differentiation of iPSCs can be used to obtain lung organoids that morphologically reflect the target tissue of the human lung very well, especially with regard to the cellular composition. The different cellular fates were investigated following the genotoxic stress induced by radiation and revealed further insights in the radiation-sensitivity of the different lung cells. Finally, we provide cellular gene sets found to be induced in the different lung organoid cellular subsets after irradiation, which could be used as additional RT response and particularly senescence gene sets in future studies. CONCLUSION By establishing these free-floating LuOrgs for the investigation of cancer therapeutic approaches as a new and patient-oriented in vitro platform particularly in experimental radiooncology, not only a reduction in the number of experimental animals, but also an adequately and meaningfully replacement of corresponding animal experiments can be achieved.
Collapse
Affiliation(s)
- Bettina Budeus
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Chiara Kroepel
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Lisa Marie Stasch
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Diana Klein
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
99
|
Perampalam P, McDonald JI, Dick FA. GO-CRISPR: A highly controlled workflow to discover gene essentiality in loss-of-function screens. PLoS One 2024; 19:e0315923. [PMID: 39693330 DOI: 10.1371/journal.pone.0315923] [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: 08/08/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Genome-wide CRISPR screens are an effective discovery tool for genes that underlie diverse cellular mechanisms that can be scored through cell fitness. Loss-of-function screens are particularly challenging compared to gain-of-function because of the limited dynamic range of decreased sgRNA sequence detection. Here we describe Guide-Only control CRISPR (GO-CRISPR), an improved loss-of-function screening workflow, and its companion software package, Toolset for the Ranked Analysis of GO-CRISPR Screens (TRACS). We demonstrate a typical GO-CRISPR workflow in a non-proliferative 3D spheroid model of dormant high grade serous ovarian cancer and demonstrate superior performance to standard screening methods. The unique integration of the pooled sgRNA library quality and guide-only controls allows TRACS to identify novel molecular pathways that were previously unidentified in tumor dormancy and undetectable to analysis packages that lack the guide only controls. Together, GO-CRISPR and TRACS can robustly improve the discovery of essential genes in challenging biological scenarios such as growth arrested cells.
Collapse
Affiliation(s)
- Pirunthan Perampalam
- London Health Sciences Centre Research Institute, London Regional Cancer Program, London, ON, Canada
- Department of Biochemistry, Western University, London, ON, Canada
- Copoly.ai Inc., Ottawa, ON, Canada
| | - James I McDonald
- London Health Sciences Centre Research Institute, London Regional Cancer Program, London, ON, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Frederick A Dick
- London Health Sciences Centre Research Institute, London Regional Cancer Program, London, ON, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
| |
Collapse
|
100
|
Yan X, Tan D, Yu L, Li D, Huang W, Huang W, Wu H. A High-Throughput and Logarithm-Serial-Dilution Microfluidic Chip for Combinational Drug Screening on Tumor Organoids. ACS Pharmacol Transl Sci 2024; 7:4135-4143. [PMID: 39698291 PMCID: PMC11650729 DOI: 10.1021/acsptsci.4c00565] [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: 09/23/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024]
Abstract
Tumor organoids are biological models for studying precision medicine. Microfluidic technology offers significant benefits for high throughput drug screening using tumor organoids. However, the range of concentrations achievable with traditional linear gradient generators in microfluidics is restricted, generating logarithmic drug concentration gradients by adjusting the channel ratio in the chip is confined to single-drug dilution chips, significantly restricting the application of microfluidics in drug screening. Here, we presented a microfluidic chip featuring continuous dilution capabilities, which generates logarithmic stepwise drug concentration gradients. We have devised a "mathematical-circuit-chip" model for designing such chips, and based on this model, we have developed and fabricated a device capable of providing 36 distinct drug concentration conditions for two types of drugs. The chip is composed of two structurally identical yet orthogonally arranged layers, each containing a dilution network capable of forming a 5-fold gradient and a tumor organoid culture module. Drug and culture medium delivery to the open culture chamber array is driven by syringe pumps. We have conducted drug screening experiments on patient-derived tumor organoids. This device facilitates high-throughput drug screening for patient-derived organoids, representing a significant stride toward the realization of precision medicine.
Collapse
Affiliation(s)
- Xingyang Yan
- Department
of Chemistry, The Hong Kong University of
Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, China
| | - Deng Tan
- Department
of Chemistry, The Hong Kong University of
Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, China
- Department
of Biology, Southern University of Science
and Technology, Shenzhen 518055, Guangdong, China
| | - Lei Yu
- Shenzhen
Institute of Synthetic Biology, Shenzhen
Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - DanYu Li
- Department
of Chemistry, The Hong Kong University of
Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, China
| | - Wei Huang
- Department
of Biology, Southern University of Science
and Technology, Shenzhen 518055, Guangdong, China
| | - Weiren Huang
- Shenzhen
Institute of Synthetic Biology, Shenzhen
Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department
of Urology, Shenzhen Institute of Translational Medicine, Shenzhen
Institutes of Advanced Technology, The First
Affiliated Hospital of Shenzhen University, International Cancer Center
of Shenzhen University, Shenzhen 518039, China
| | - Hongkai Wu
- Department
of Chemistry, The Hong Kong University of
Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, China
- The
Hong Kong University of Science and Technology Shenzhen Research Institute, Shenzhen 518172, China
| |
Collapse
|