1
|
Xu X, Guo S, Gu H, Cha Z, Shi X, Yin X, Wang H, Gao S, Li B, Zhu L, Jing W, Zheng K, Shao Z, Cheng P, Zheng C, Shih YP, Li Y, Qian B, Gao D, Tran E, Jin G. Identification and validation of a T cell receptor targeting KRAS G12V in HLA-A*11:01 pancreatic cancer patients. JCI Insight 2025; 10:e181873. [PMID: 39846249 DOI: 10.1172/jci.insight.181873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025] Open
Abstract
T cells targeting a KRAS mutation can induce durable tumor regression in some patients with metastatic epithelial cancer. It is unknown whether T cells targeting mutant KRAS that are capable of killing tumor cells can be identified from peripheral blood of patients with pancreatic cancer. We developed an in vitro stimulation approach and identified HLA-A*11:01-restricted KRAS G12V-reactive CD8+ T cells and HLA-DRB1*15:01-restricted KRAS G12V-reactive CD4+ T cells from peripheral blood of 2 out of 6 HLA-A*11:01-positive patients with pancreatic cancer whose tumors expressed KRAS G12V. The HLA-A*11:01-restricted KRAS G12V-reactive T cell receptor (TCR) was isolated and validated to specifically recognize the KRAS G12V8-16 neoepitope. While T cells engineered to express this TCR specifically recognized all 5 tested human HLA-A*11:01+ and KRAS G12V+ pancreatic cancer organoids, the recognition was often modest, and tumor cell killing was observed in only 2 out of 5 organoids. IFN-γ priming of the organoids enhanced the recognition and killing by the TCR-engineered T cells. The TCR-engineered T cells could significantly slow the growth of an established organoid-derived xenograft in immunodeficient mice. Our data suggest that this TCR has potential for use in TCR-gene therapy, but additional strategies that enhance tumor recognition by the TCR-engineered T cells likely will be required to increase clinical activity.
Collapse
Affiliation(s)
- Xiongfei Xu
- Department of Hepatobiliary Pancreatic Surgery
- Shanghai Institute of Pancreatic Diseases, and
| | - Shiwei Guo
- Department of Hepatobiliary Pancreatic Surgery
- Shanghai Institute of Pancreatic Diseases, and
| | - Haihui Gu
- Department of Transfusion Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhanshan Cha
- Department of Transfusion Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery
| | - Xiaoyi Yin
- Department of Hepatobiliary Pancreatic Surgery
| | - Huan Wang
- Department of Hepatobiliary Pancreatic Surgery
| | - Suizhi Gao
- Department of Hepatobiliary Pancreatic Surgery
| | - Bo Li
- Department of Hepatobiliary Pancreatic Surgery
| | - Lingyu Zhu
- Department of Hepatobiliary Pancreatic Surgery
| | - Wei Jing
- Department of Hepatobiliary Pancreatic Surgery
| | | | - Zhuo Shao
- Department of Hepatobiliary Pancreatic Surgery
| | - Peng Cheng
- Department of Hepatobiliary Pancreatic Surgery
| | - Chunhong Zheng
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
- International Cancer Institute, Peking University, Beijing, China
| | - Yi-Ping Shih
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Yunguang Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Baohua Qian
- Department of Transfusion Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dong Gao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Eric Tran
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery
- Shanghai Institute of Pancreatic Diseases, and
| |
Collapse
|
2
|
Xu J, Pham MD, Corbo V, Ponz-Sarvise M, Oni T, Öhlund D, Hwang CI. Advancing pancreatic cancer research and therapeutics: the transformative role of organoid technology. Exp Mol Med 2025:10.1038/s12276-024-01378-w. [PMID: 39814914 DOI: 10.1038/s12276-024-01378-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 01/18/2025] Open
Abstract
Research on pancreatic cancer has transformed with the advent of organoid technology, providing a better platform that closely mimics cancer biology in vivo. This review highlights the critical advancements facilitated by pancreatic organoid models in understanding disease progression, evaluating therapeutic responses, and identifying biomarkers. These three-dimensional cultures enable the proper recapitulation of the cellular architecture and genetic makeup of the original tumors, providing insights into the complex molecular and cellular dynamics at various stages of pancreatic ductal adenocarcinoma (PDAC). We explore the applications of pancreatic organoids in dissecting the tumor microenvironment (TME); elucidating cancer progression, metastasis, and drug resistance mechanisms; and personalizing therapeutic strategies. By overcoming the limitations of traditional 2D cultures and animal models, the use of pancreatic organoids has significantly accelerated translational research, which is promising for improving diagnostic and therapeutic approaches in clinical settings, ultimately aiming to improve the outcomes of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Jihao Xu
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Minh Duc Pham
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Mariano Ponz-Sarvise
- Department of Medical Oncology and Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Pamplona, Spain
| | - Tobiloba Oni
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Daniel Öhlund
- Umeå University, Department of Diagnostics and Intervention, and Wallenberg Centre for Molecular Medicine at Umeå University, Umeå, Sweden
| | - Chang-Il Hwang
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA.
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
3
|
Wang Z, Zhao F, Lang H, Ren H, Zhang Q, Huang X, He C, Xu C, Tan C, Ma J, Duan S, Wang Z. Organoids in skin wound healing. BURNS & TRAUMA 2025; 13:tkae077. [PMID: 39759541 PMCID: PMC11697111 DOI: 10.1093/burnst/tkae077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/30/2024] [Accepted: 11/11/2024] [Indexed: 01/07/2025]
Abstract
Stem cells (SCs) can self-replicate and differentiate into multiple lineages. Organoids, 3D cultures derived from SCs, can replicate the spatial structure and physiological characteristics of organs in vitro. Skin organoids can effectively simulate the physiological structure and function of skin tissue, reliably restoring the natural skin ecology in various in vitro environments. Skin organoids have been employed extensively in skin development and pathology research, offering valuable insights for drug screening. Moreover, they play crucial roles in skin regeneration and tissue repair. This in-depth review explores the construction and applications of skin organoids in wound healing, with a focus on their construction process, including skin appendage integration, and significant advancements in wound-healing research.
Collapse
Affiliation(s)
- Zitong Wang
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, No. 77 Puhe Road, Shenyang, Liaoning 110013, China
| | - Hongxin Lang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, No. 77 Puhe Road, Shenyang, Liaoning 110013, China
| | - Haiyue Ren
- Department of Pathology, Wuhan Hospital of Traditional Chinese and Western Medicine (Wuhan No. 1 Hospital), No. 215 Zhongshan Street, Wuhan, Hubei 430022, China
| | - Qiqi Zhang
- Department of Pathology, Chengdu Third People's Hospital, No. 82 Qinglong Street, Chengdu, Sichuan 610031, China
| | - Xing Huang
- Department of Anaesthesiology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yantaxi Road, Xi'an, Shanxi 710061, China
| | - Cai He
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Chengcheng Xu
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Chiyu Tan
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Jiajie Ma
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Shu Duan
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Zhe Wang
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| |
Collapse
|
4
|
Huang P, Gao W, Fu C, Wang M, Li Y, Chu B, He A, Li Y, Deng X, Zhang Y, Kong Q, Yuan J, Wang H, Shi Y, Gao D, Qin R, Hunter T, Tian R. Clinical functional proteomics of intercellular signalling in pancreatic cancer. Nature 2025; 637:726-735. [PMID: 39537929 DOI: 10.1038/s41586-024-08225-y] [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: 07/21/2023] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an atypical, highly stromal tumour microenvironment (TME) that profoundly contributes to its poor prognosis1. Here, to better understand the intercellular signalling between cancer and stromal cells directly in PDAC tumours, we developed a multidimensional proteomic strategy called TMEPro. We applied TMEPro to profile the glycosylated secreted and plasma membrane proteome of 100 human pancreatic tissue samples to a great depth, define cell type origins and identify potential paracrine cross-talk, especially that mediated through tyrosine phosphorylation. Temporal dynamics during pancreatic tumour progression were investigated in a genetically engineered PDAC mouse model. Functionally, we revealed reciprocal signalling between stromal cells and cancer cells mediated by the stromal PDGFR-PTPN11-FOS signalling axis. Furthermore, we examined the generic shedding mechanism of plasma membrane proteins in PDAC tumours and revealed that matrix-metalloprotease-mediated shedding of the AXL receptor tyrosine kinase ectodomain provides an additional dimension of intercellular signalling regulation in the PDAC TME. Importantly, the level of shed AXL has a potential correlation with lymph node metastasis, and inhibition of AXL shedding and its kinase activity showed a substantial synergistic effect in inhibiting cancer cell growth. In summary, we provide TMEPro, a generically applicable clinical functional proteomic strategy, and a comprehensive resource for better understanding the PDAC TME and facilitating the discovery of new diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Peiwu Huang
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Weina Gao
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Changying Fu
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunguang Li
- Key Laboratory of Multi-Cell Systems, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Bizhu Chu
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - An He
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Yuan Li
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Xiaomei Deng
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Yehan Zhang
- Key Laboratory of Multi-Cell Systems, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Qian Kong
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Jingxiong Yuan
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hebin Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Shi
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
- Bristol Myers Squibb, San Diego, CA, USA.
| | - Dong Gao
- Key Laboratory of Multi-Cell Systems, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China.
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ruijun Tian
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
5
|
Acimovic I, Gabrielová V, Martínková S, Eid M, Vlažný J, Moravčík P, Hlavsa J, Moráň L, Cakmakci RC, Staňo P, Procházka V, Kala Z, Trnka J, Vaňhara P. Ex-Vivo 3D Cellular Models of Pancreatic Ductal Adenocarcinoma: From Embryonic Development to Precision Oncology. Pancreas 2025; 54:e57-e71. [PMID: 39074056 DOI: 10.1097/mpa.0000000000002393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
ABSTRACT Pancreas is a vital gland of gastrointestinal system with exocrine and endocrine secretory functions, interweaved into essential metabolic circuitries of the human body. Pancreatic ductal adenocarcinoma (PDAC) represents one of the most lethal malignancies, with a 5-year survival rate of 11%. This poor prognosis is primarily attributed to the absence of early symptoms, rapid metastatic dissemination, and the limited efficacy of current therapeutic interventions. Despite recent advancements in understanding the etiopathogenesis and treatment of PDAC, there remains a pressing need for improved individualized models, identification of novel molecular targets, and development of unbiased predictors of disease progression. Here we aim to explore the concept of precision medicine utilizing 3-dimensional, patient-specific cellular models of pancreatic tumors and discuss their potential applications in uncovering novel druggable molecular targets and predicting clinical parameters for individual patients.
Collapse
Affiliation(s)
- Ivana Acimovic
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Viktorie Gabrielová
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Stanislava Martínková
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague
| | - Michal Eid
- Departments of Internal Medicine, Hematology and Oncology
| | | | - Petr Moravčík
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Jan Hlavsa
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | | | - Riza Can Cakmakci
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Peter Staňo
- Departments of Internal Medicine, Hematology and Oncology
| | - Vladimír Procházka
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Zdeněk Kala
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Jan Trnka
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague
| | | |
Collapse
|
6
|
Kim B, Park J, Na HY, Park S, Jin J, Jung K, Lee JC, Hwang JH, Seo M, Kim J. The origin of patient-derived cancer organoids from pathologically undiagnosed specimen in patients with pancreatobiliary cancers. Cell Oncol (Dordr) 2024:10.1007/s13402-024-01026-5. [PMID: 39688793 DOI: 10.1007/s13402-024-01026-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
PURPOSE Tissue confirmation of pancreatobiliary cancer is often difficult because of the location of the tumor and structure of the surrounding blood vessels. Patient-derived cancer organoids (PDCOs) reflect the genomic characteristics of individual cancers. Although diverse attempts to construct PDCOs for various pancreatobiliary cancer models are ongoing, no research results have yet confirmed the possibility of performing a precise diagnosis on PDCOs derived from pathologically negative patient samples. METHODS We obtained a total of nine samples, including pathologically negative samples, from four patients (three patients with pancreatic cancer and one patient with gallbladder cancer) using different tissue acquisition methods to establish PDCOs (success rate 75%). RESULTS We successfully verified whether the constructed PDCOs could represent the tissues of patients with pancreatobiliary cancer at each multi-omics level using tumor panel sequencing, single-cell RNA sequencing, hematoxylin and eosin, and immunohistochemical staining. PDCOs from pathologically negative samples showed expression patterns of malignant ductal cell-related biomarkers similar to those of other pathologically positive samples. Furthermore, the expression patterns at the single-cell level in PDCO from patients ultimately diagnosed with gallbladder cancer after surgery were different from those in patients with pancreatic cancer. CONCLUSION Therefore, our study implicated the potential of PDCOs as diagnostic and research tools, including for case involving limited tissue samples. Based on these results, we anticipate that this could be extended to more advanced studies, such as drug sensitivity testing, through large-scale trials in the near future.
Collapse
Affiliation(s)
- Bomi Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | | | - Hee Young Na
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Sinwoo Park
- Department of Computer and Information Science, Korea University, Sejong, Korea
| | | | - Kwangrok Jung
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jong-Chan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Minseok Seo
- Department of Computer and Information Science, Korea University, Sejong, Korea.
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea.
| |
Collapse
|
7
|
Zhang Y, Qi F, Chen P, Liu BF, Li Y. Spatially defined microenvironment for engineering organoids. BIOPHYSICS REVIEWS 2024; 5:041302. [PMID: 39679203 PMCID: PMC11646138 DOI: 10.1063/5.0198848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 10/01/2024] [Indexed: 12/17/2024]
Abstract
In the intricately defined spatial microenvironment, a single fertilized egg remarkably develops into a conserved and well-organized multicellular organism. This observation leads us to hypothesize that stem cells or other seed cell types have the potential to construct fully structured and functional tissues or organs, provided the spatial cues are appropriately configured. Current organoid technology, however, largely depends on spontaneous growth and self-organization, lacking systematic guided intervention. As a result, the structures replicated in vitro often emerge in a disordered and sparse manner during growth phases. Although existing organoids have made significant contributions in many aspects, such as advancing our understanding of development and pathogenesis, aiding personalized drug selection, as well as expediting drug development, their potential in creating large-scale implantable tissue or organ constructs, and constructing multicomponent microphysiological systems, together with functioning at metabolic levels remains underutilized. Recent discoveries have demonstrated that the spatial definition of growth factors not only induces directional growth and migration of organoids but also leads to the formation of assembloids with multiple regional identities. This opens new avenues for the innovative engineering of higher-order organoids. Concurrently, the spatial organization of other microenvironmental cues, such as physical stresses, mechanical loads, and material composition, has been minimally explored. This review delves into the burgeoning field of organoid engineering with a focus on potential spatial microenvironmental control. It offers insight into the molecular principles, expected outcomes, and potential applications, envisioning a future perspective in this domain.
Collapse
Affiliation(s)
- Yilan Zhang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fukang Qi
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
8
|
Zeng G, Yu Y, Wang M, Liu J, He G, Yu S, Yan H, Yang L, Li H, Peng X. Advancing cancer research through organoid technology. J Transl Med 2024; 22:1007. [PMID: 39516934 PMCID: PMC11545094 DOI: 10.1186/s12967-024-05824-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
The complexity of tumors and the challenges associated with treatment often stem from the limitations of existing models in accurately replicating authentic tumors. Recently, organoid technology has emerged as an innovative platform for tumor research. This bioengineering approach enables researchers to simulate, in vitro, the interactions between tumors and their microenvironment, thereby enhancing the intricate interplay between tumor cells and their surroundings. Organoids also integrate multidimensional data, providing a novel paradigm for understanding tumor development and progression while facilitating precision therapy. Furthermore, advancements in imaging and genetic editing techniques have significantly augmented the potential of organoids in tumor research. This review explores the application of organoid technology for more precise tumor simulations and its specific contributions to cancer research advancements. Additionally, we discuss the challenges and evolving trends in developing comprehensive tumor models utilizing organoid technology.
Collapse
Affiliation(s)
- Guolong Zeng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Yifan Yu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Meiting Wang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Jiaxing Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Guangpeng He
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Sixuan Yu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Huining Yan
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, China.
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, China.
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, China.
| |
Collapse
|
9
|
Kim SC, Seo HY, Lee JO, Maeng JE, Shin YK, Lee SH, Jang JY, Ku JL. Establishment, characterization, and biobanking of 36 pancreatic cancer organoids: prediction of metastasis in resectable pancreatic cancer. Cell Oncol (Dordr) 2024; 47:1627-1647. [PMID: 38619751 PMCID: PMC11467084 DOI: 10.1007/s13402-024-00939-5] [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: 03/13/2024] [Indexed: 04/16/2024] Open
Abstract
PURPOSE Early dissemination of primary pancreatic ductal adenocarcinoma (PDAC) is the main cause of dismal prognosis as it highly limits possible treatment options. A number of PDAC patients experience distant metastasis even after treatment due to the metastatic clones. We aimed to demonstrate the molecular architecture of borderline resectable PDAC manifests cancer dissemination of PDAC. METHODS Here, 36 organoids isolated from primary tumor masses of PDAC patients with diverse metastatic statues are presented. Whole-exome sequencing and RNA sequencing were performed and drug responses to clinically relevant 18 compounds were assessed. RESULTS Our results revealed that borderline resectable PDAC organoids exhibited distinct patterns according to their metastatic potency highlighted by multiple genetic and transcriptional factors and strong variances in drug responses. CONCLUSIONS These data suggest that the presence of metastatic PDAC can be identified by integrating molecular compositions and drug responses of borderline resectable PDAC.
Collapse
Affiliation(s)
- Soon-Chan Kim
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ha-Young Seo
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Ja-Oh Lee
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Ju Eun Maeng
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Young-Kyoung Shin
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Sang Hyub Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| | - Jin-Young Jang
- Department of Surgery, Seoul National University College of Medicine, 103, Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
| | - Ja-Lok Ku
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
10
|
Grützmeier SE, Sodal HMM, Kovacevic B, Vilmann P, Karstensen JG, Klausen P. EUS-guided biopsies versus surgical specimens for establishing patient-derived pancreatic cancer organoids: a systematic review and meta-analysis. Gastrointest Endosc 2024; 100:750-755. [PMID: 38593932 DOI: 10.1016/j.gie.2024.04.012] [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] [Received: 08/29/2023] [Revised: 02/22/2024] [Accepted: 04/01/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND AND AIMS Patient-derived tumor organoids (PDTOs) are a promising new disease model in pancreatic cancer for use in personalized medicine. However, the overall success rate (SR) of establishing these cultures from EUS-guided biopsies is unknown. METHODS We searched relevant database publications reporting SRs of PDTO establishment from pancreatic cancer. The primary outcome was SR stratified on tissue acquisition method (EUS-guided biopsies, percutaneous biopsies, and surgical specimens). RESULTS Twenty-four studies were identified that included 1053 attempts at establishing PDTOs. Overall SR was 63% (95% confidence interval [CI], 54%-72%). Pooled SRs of PDTO establishment from EUS-guided biopsies, percutaneous biopsies, and surgical specimens were 60% (95% CI, 43%-76%), 36% (95% CI, 14%-61%), and 62% (95% CI, 48%-75%), respectively, and did not differ significantly (P = .1975). CONCLUSION The SR of PDTO establishment from EUS-guided biopsies is comparable to that from surgical specimens. Both techniques are suitable for tissue acquisition for PDTOs in clinical and research settings. (PROSPERO registration number: CRD42023425121.).
Collapse
Affiliation(s)
- Simon Ezban Grützmeier
- Gastro Unit, Endoscopic Division, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark.
| | - Hafsa Mahad Mahamud Sodal
- Gastro Unit, Endoscopic Division, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark
| | - Bojan Kovacevic
- Gastro Unit, Endoscopic Division, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark; Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Peter Vilmann
- Gastro Unit, Endoscopic Division, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - John Gásdal Karstensen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Pancreatitis Centre East, Gastro Unit, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Pia Klausen
- Gastro Unit, Endoscopic Division, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark; Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
11
|
Tamagawa H, Fujii M, Togasaki K, Seino T, Kawasaki S, Takano A, Toshimitsu K, Takahashi S, Ohta Y, Matano M, Kawasaki K, Machida Y, Sekine S, Machinaga A, Sasai K, Kodama Y, Kakiuchi N, Ogawa S, Hirano T, Seno H, Kitago M, Kitagawa Y, Iwasaki E, Kanai T, Sato T. Wnt-deficient and hypoxic environment orchestrates squamous reprogramming of human pancreatic ductal adenocarcinoma. Nat Cell Biol 2024; 26:1759-1772. [PMID: 39232216 DOI: 10.1038/s41556-024-01498-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 08/05/2024] [Indexed: 09/06/2024]
Abstract
Human pancreatic cancer is characterized by the molecular diversity encompassing native duct-like and squamous cell-like identities, but mechanisms underlying squamous transdifferentiation have remained elusive. To comprehensively capture the molecular diversity of human pancreatic cancer, we here profiled 65 patient-derived pancreatic cancer organoid lines, including six adenosquamous carcinoma lines. H3K27me3-mediated erasure of the ductal lineage specifiers and hijacking of the TP63-driven squamous-cell programme drove squamous-cell commitment, providing survival benefit in a Wnt-deficient environment and hypoxic conditions. Gene engineering of normal pancreatic duct organoids revealed that GATA6 loss and a Wnt-deficient environment, in concert with genetic or hypoxia-mediated inactivation of KDM6A, facilitate squamous reprogramming, which in turn enhances environmental fitness. EZH2 inhibition counterbalanced the epigenetic bias and curbed the growth of adenosquamous cancer organoids. Our results demonstrate how an adversarial microenvironment dictates the molecular and histological evolution of human pancreatic cancer and provide insights into the principles and significance of lineage conversion in human cancer.
Collapse
Affiliation(s)
- Hiroki Tamagawa
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Fujii
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan.
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
| | - Kazuhiro Togasaki
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Seino
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Shintaro Kawasaki
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Ai Takano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Kohta Toshimitsu
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Sirirat Takahashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Ohta
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mami Matano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Kenta Kawasaki
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Yujiro Machida
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Shigeki Sekine
- Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | | | | | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Nobuyuki Kakiuchi
- Department of Pathology and Tumor Biology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomonori Hirano
- Department of Pathology and Tumor Biology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Eisuke Iwasaki
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Takanori Kanai
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan.
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
12
|
Sun H, Wang Y, Sun M, Ke X, Li C, Jin B, Pang M, Wang Y, Jiang S, Du L, Du S, Zhong S, Zhao H, Pang Y, Sun Y, Yang Z, Yang H, Mao Y. Developing Patient-Derived 3D-Bioprinting models of pancreatic cancer. J Adv Res 2024:S2090-1232(24)00413-2. [PMID: 39278567 DOI: 10.1016/j.jare.2024.09.011] [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: 06/11/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024] Open
Abstract
INTRODUCTION Pancreatic cancer (PC) remains a challenging malignancy, and adjuvant chemotherapy is critical in improving patient survival post-surgery. However, the intrinsic heterogeneity of PC necessitates personalized treatment strategies, highlighting the need for reliable preclinical models. OBJECTIVES This study aimed to develop novel patient-derived preclinical PC models using three-dimensional bioprinting (3DP) technology. METHODS Patient-derived PC models were established using 3DP technology. Genomic and histological analyses were performed to characterize these models and compare them with corresponding patient tissues. Chemotherapeutic drug sensitivity tests were conducted on the PC 3DP models, and correlations with clinical outcomes were analyzed. RESULTS The study successfully established PC 3DP models with a modeling success rate of 86.96%. These models preserved genomic and histological features consistent with patient tissues. Drug sensitivity testing revealed significant heterogeneity among PC 3DP models, mirroring clinical variability, and potential correlations with clinical outcomes. CONCLUSION The PC 3DP models demonstrated their utility as reliable preclinical tools, retaining key genomic and histological characteristics. Importantly, drug sensitivity profiles in these models showed potential correlations with clinical outcomes, indicating their promise in customizing treatment strategies and predicting patient prognoses. Further validation with larger patient cohorts is warranted to confirm their potential clinical utility.
Collapse
Affiliation(s)
- Hang Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China; Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, 100730, China
| | - Yan Wang
- Eight-year MD Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Minghao Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Xindi Ke
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China; Eight-year MD Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Changcan Li
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Bao Jin
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Mingchang Pang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Yanan Wang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Shangze Jiang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Liwei Du
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Shouxian Zhong
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Yuan Pang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China; Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, 100084, China
| | - Yongliang Sun
- First Department of Hepatopancreatobiliary Surgery, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Zhiying Yang
- First Department of Hepatopancreatobiliary Surgery, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China.
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China.
| |
Collapse
|
13
|
de Back TR, van Hooff SR, Sommeijer DW, Vermeulen L. Transcriptomic subtyping of gastrointestinal malignancies. Trends Cancer 2024; 10:842-856. [PMID: 39019673 DOI: 10.1016/j.trecan.2024.06.007] [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: 04/25/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/19/2024]
Abstract
Gastrointestinal (GI) cancers are highly heterogeneous at multiple levels. Tumor heterogeneity can be captured by molecular profiling, such as genetic, epigenetic, proteomic, and transcriptomic classification. Transcriptomic subtyping has the advantage of combining genetic and epigenetic information, cancer cell-intrinsic properties, and the tumor microenvironment (TME). Unsupervised transcriptomic subtyping systems of different GI malignancies have gained interest because they reveal shared biological features across cancers and bear prognostic and predictive value. Importantly, transcriptomic subtypes accurately reflect complex phenotypic states varying not only per tumor region, but also throughout disease progression, with consequences for clinical management. Here, we discuss methodologies of transcriptomic subtyping, proposed taxonomies for GI malignancies, and the challenges posed to clinical implementation, highlighting opportunities for future transcriptomic profiling efforts to optimize clinical impact.
Collapse
Affiliation(s)
- Tim R de Back
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Sander R van Hooff
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Dirkje W Sommeijer
- Flevohospital, Department of Internal Medicine, Hospitaalweg 1, 1315 RA, Almere, The Netherlands
| | - Louis Vermeulen
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
Wang Y, Ye Z, Lou X, Xu J, Jing D, Zhou C, Qin Y, Chen J, Xu X, Yu X, Ji S. Comparison among different preclinical models derived from the same patient with a non-functional pancreatic neuroendocrine tumor. Hum Cell 2024; 37:1522-1534. [PMID: 39078546 DOI: 10.1007/s13577-024-01107-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024]
Abstract
Pancreatic neuroendocrine tumors are the second most common tumors of the pancreas, and approximately half of patients are diagnosed with liver metastases. Currently, the improvement in the efficacy of relevant treatment methods is still limited. Therefore, there is an urgent need for in-depth research on the molecular biological mechanism of pancreatic neuroendocrine tumors. However, due to their relatively inert biology, preclinical models are extremely scarce. Here, the patient-derived organoid, and patient-derived xenograft were successfully constructed. These two models and the previously constructed cell line named SPNE1 all derived from the same patient with a grade 3 non-functional pancreatic neuroendocrine tumor, providing new tumor modeling platforms, and characterized using immunohistochemistry, whole-exome sequencing, and single-cell transcriptome sequencing. Combined with a tumor formation experiment in immunodeficient mice, we selected the model that most closely recapitulated the parental tumor. Overall, the patient-derived xenograft model most closely resembled human tumor tissue.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xin Lou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Junfeng Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Desheng Jing
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chenjie Zhou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jie Chen
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Head and Neck and Neuroendocrine Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
15
|
Hu D, Zhao T, Xu C, Pan X, Zhou Z, Wang S. Epigenetic Modifiers in Cancer Metastasis. Biomolecules 2024; 14:916. [PMID: 39199304 PMCID: PMC11352731 DOI: 10.3390/biom14080916] [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: 06/03/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
Metastasis is the primary cause of cancer-related death, with the dissemination and colonization of primary tumor cells at the metastatic site facilitated by various molecules and complex pathways. Understanding the biological mechanisms underlying the metastatic process is critical for the development of effective interventions. Several epigenetic modifications have been identified that play critical roles in regulating cancer metastasis. This review aims to provide a comprehensive summary of recent advances in understanding the role of epigenetic modifiers, including histone modifications, DNA methylation, non-coding RNAs, enhancer reprogramming, chromatin accessibility, and N6-methyladenosine, in metastasis-associated processes, such as epithelial-mesenchymal transition (EMT), cancer cell migration, and invasion. In particular, this review provides a detailed and in-depth description of the role of crosstalk between epigenetic regulators in tumor metastasis. Additionally, we explored the potential and limitations of epigenetics-related target molecules in the diagnosis, treatment, and prognosis of cancer metastasis.
Collapse
Affiliation(s)
- Die Hu
- Key Laboratory of Molecular Genetics between Kangda College of Nanjing Medical University and Suzhou Medical College of Soochow University, Suzhou 215123, China;
- Department of Basic Medicine, Kangda College of Nanjing Medical University, Lianyungang 222000, China; (C.X.); (X.P.)
| | - Tianci Zhao
- Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, China;
| | - Chenxing Xu
- Department of Basic Medicine, Kangda College of Nanjing Medical University, Lianyungang 222000, China; (C.X.); (X.P.)
| | - Xinyi Pan
- Department of Basic Medicine, Kangda College of Nanjing Medical University, Lianyungang 222000, China; (C.X.); (X.P.)
| | - Zhengyu Zhou
- Key Laboratory of Molecular Genetics between Kangda College of Nanjing Medical University and Suzhou Medical College of Soochow University, Suzhou 215123, China;
- Laboratory Animal Center, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Shengjie Wang
- Key Laboratory of Molecular Genetics between Kangda College of Nanjing Medical University and Suzhou Medical College of Soochow University, Suzhou 215123, China;
- Department of Basic Medicine, Kangda College of Nanjing Medical University, Lianyungang 222000, China; (C.X.); (X.P.)
- Laboratory Animal Center, Suzhou Medical College of Soochow University, Suzhou 215123, China
| |
Collapse
|
16
|
Fu Y, Tao J, Liu T, Liu Y, Qiu J, Su D, Wang R, Luo W, Cao Z, Weng G, Zhang T, Zhao Y. Unbiasedly decoding the tumor microenvironment with single-cell multiomics analysis in pancreatic cancer. Mol Cancer 2024; 23:140. [PMID: 38982491 PMCID: PMC11232163 DOI: 10.1186/s12943-024-02050-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with a poor prognosis and limited therapeutic options. Research on the tumor microenvironment (TME) of PDAC has propelled the development of immunotherapeutic and targeted therapeutic strategies with a promising future. The emergence of single-cell sequencing and mass spectrometry technologies, coupled with spatial omics, has collectively revealed the heterogeneity of the TME from a multiomics perspective, outlined the development trajectories of cell lineages, and revealed important functions of previously underrated myeloid cells and tumor stroma cells. Concurrently, these findings necessitated more refined annotations of biological functions at the cell cluster or single-cell level. Precise identification of all cell clusters is urgently needed to determine whether they have been investigated adequately and to identify target cell clusters with antitumor potential, design compatible treatment strategies, and determine treatment resistance. Here, we summarize recent research on the PDAC TME at the single-cell multiomics level, with an unbiased focus on the functions and potential classification bases of every cellular component within the TME, and look forward to the prospects of integrating single-cell multiomics data and retrospectively reusing bulk sequencing data, hoping to provide new insights into the PDAC TME.
Collapse
Affiliation(s)
- Yifan Fu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jinxin Tao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Tao Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yueze Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jiangdong Qiu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Dan Su
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ruobing Wang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Wenhao Luo
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhe Cao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guihu Weng
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
17
|
Thorel L, Perréard M, Florent R, Divoux J, Coffy S, Vincent A, Gaggioli C, Guasch G, Gidrol X, Weiswald LB, Poulain L. Patient-derived tumor organoids: a new avenue for preclinical research and precision medicine in oncology. Exp Mol Med 2024; 56:1531-1551. [PMID: 38945959 PMCID: PMC11297165 DOI: 10.1038/s12276-024-01272-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 03/18/2024] [Accepted: 04/14/2024] [Indexed: 07/02/2024] Open
Abstract
Over the past decade, the emergence of patient-derived tumor organoids (PDTOs) has broadened the repertoire of preclinical models and progressively revolutionized three-dimensional cell culture in oncology. PDTO can be grown from patient tumor samples with high efficiency and faithfully recapitulates the histological and molecular characteristics of the original tumor. Therefore, PDTOs can serve as invaluable tools in oncology research, and their translation to clinical practice is exciting for the future of precision medicine in oncology. In this review, we provide an overview of methods for establishing PDTOs and their various applications in cancer research, starting with basic research and ending with the identification of new targets and preclinical validation of new anticancer compounds and precision medicine. Finally, we highlight the challenges associated with the clinical implementation of PDTO, such as its representativeness, success rate, assay speed, and lack of a tumor microenvironment. Technological developments and autologous cocultures of PDTOs and stromal cells are currently ongoing to meet these challenges and optimally exploit the full potential of these models. The use of PDTOs as standard tools in clinical oncology could lead to a new era of precision oncology in the coming decade.
Collapse
Grants
- AP-RM-19-020 Fondation de l'Avenir pour la Recherche Médicale Appliquée (Fondation de l'Avenir)
- PJA20191209649 Fondation ARC pour la Recherche sur le Cancer (ARC Foundation for Cancer Research)
- TRANSPARANCE Fondation ARC pour la Recherche sur le Cancer (ARC Foundation for Cancer Research)
- TRANSPARANCE Ligue Contre le Cancer
- ORGAPRED Ligue Contre le Cancer
- 3D-Hub Canceropôle PACA (Canceropole PACA)
- Pré-néo 2019-188 Institut National Du Cancer (French National Cancer Institute)
- Conseil Régional de Haute Normandie (Upper Normandy Regional Council)
- GIS IBiSA, Cancéropôle Nord-Ouest (ORGRAFT project), the Groupement des Entreprises Françaises dans la Lutte contre le Cancer (ORGAVADS project), the Fonds de dotation Patrick de Brou de Laurière (ORGAVADS project),and Normandy County Council (ORGATHEREX project).
- GIS IBiSA, Cancéropôle Nord-Ouest (OrgaNO project), Etat-région
- GIS IBiSA, Region Sud
- GIS IBiSA, Cancéropôle Nord-Ouest (OrgaNO project), and Normandy County Council (ORGAPRED, PLATONUS ONE, POLARIS, and EQUIP’INNOV projects).
Collapse
Affiliation(s)
- Lucie Thorel
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Marion Perréard
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Department of Head and Neck Surgery, Caen University Hospital, Caen, France
| | - Romane Florent
- ORGAPRED core facility, US PLATON, Université de Caen Normandie, Caen, France
| | - Jordane Divoux
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
- ORGAPRED core facility, US PLATON, Université de Caen Normandie, Caen, France
| | - Sophia Coffy
- Biomics, CEA, Inserm, IRIG, UA13 BGE, Univ. Grenoble Alpes, Grenoble, France
| | - Audrey Vincent
- CNRS UMR9020, INSERM U1277, CANTHER Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, CNRS, Inserm, CHU Lille, Lille, France
| | - Cédric Gaggioli
- CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), 3D-Hub-S Facility, CNRS University Côte d'Azur, Nice, France
| | - Géraldine Guasch
- CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Epithelial Stem Cells and Cancer Team, Aix-Marseille University, Marseille, France
| | - Xavier Gidrol
- Biomics, CEA, Inserm, IRIG, UA13 BGE, Univ. Grenoble Alpes, Grenoble, France
| | - Louis-Bastien Weiswald
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France.
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France.
- ORGAPRED core facility, US PLATON, Université de Caen Normandie, Caen, France.
| | - Laurent Poulain
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France.
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France.
- ORGAPRED core facility, US PLATON, Université de Caen Normandie, Caen, France.
| |
Collapse
|
18
|
Piraino F, Costa M, Meyer M, Cornish G, Ceroni C, Garnier V, Hoehnel-Ka S, Brandenberg N. Organoid models: the future companions of personalized drug development. Biofabrication 2024; 16:032009. [PMID: 38608454 DOI: 10.1088/1758-5090/ad3e30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/12/2024] [Indexed: 04/14/2024]
Abstract
High failure rates of the current drug development process are driving exemplary changes toward methodologies centered on human diseasein-vitromodeling. Organoids are self-organized tissue sub-units resembling their organ of origin and are widely acknowledged for their unique potential in recapitulating human physio-pathological mechanisms. They are transformative for human health by becoming the platform of choice to probe disease mechanisms and advance new therapies. Furthermore, the compounds' validation as therapeutics represents another point of the drug development pipeline where organoids may provide key understandings and help pharma organizations replace or reduce animal research. In this review, we focus on gastrointestinal organoid models, which are currently the most advanced organoid models in drug development. We focus on experimental validations of their value, and we propose avenues to enhance their use in drug discovery and development, as well as precision medicine and diagnostics.
Collapse
Affiliation(s)
| | - Mariana Costa
- Doppl SA, EPFL Innovation Park, Lausanne, Switzerland
| | - Marine Meyer
- Doppl SA, EPFL Innovation Park, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
19
|
Boilève A, Cartry J, Goudarzi N, Bedja S, Mathieu JRR, Bani MA, Nicolle R, Mouawia A, Bouyakoub R, Nicotra C, Ngo-Camus M, Job B, Lipson K, Boige V, Valéry M, Tarabay A, Dartigues P, Tselikas L, de Baere T, Italiano A, Cosconea S, Gelli M, Fernandez-de-Sevilla E, Annereau M, Malka D, Smolenschi C, Ducreux M, Hollebecque A, Jaulin F. Organoids for Functional Precision Medicine in Advanced Pancreatic Cancer. Gastroenterology 2024:S0016-5085(24)05027-3. [PMID: 38866343 DOI: 10.1053/j.gastro.2024.05.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 05/01/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND & AIMS Patient-derived organoids (PDOs) are promising tumor avatars that could enable ex vivo drug tests to personalize patients' treatments in the frame of functional precision oncology. However, clinical evidence remains scarce. This study aims to evaluate whether PDOs can be implemented in clinical practice to benefit patients with advanced refractory pancreatic ductal adenocarcinoma (PDAC). METHODS During 2021 to 2022, 87 patients were prospectively enrolled in an institutional review board-approved protocol. Inclusion criteria were histologically confirmed PDAC with the tumor site accessible. A panel of 25 approved antitumor therapies (chemogram) was tested and compared to patient responses to assess PDO predictive values and map the drug sensitivity landscape in PDAC. RESULTS Fifty-four PDOs were generated from 87 pretreated patients (take-on rate, 62%). The main PDO mutations were KRAS (96%), TP53 (88%), and CDKN2A/B (22%), with a 91% concordance rate with their tumor of origin. The mean turnaround time to chemogram was 6.8 weeks. In 91% of cases, ≥1 hit was identified (gemcitabine (n = 20 of 54), docetaxel (n = 18 of 54), and vinorelbine (n = 17 of 54), with a median of 3 hits/patient (range, 0-12). Our cohort included 34 evaluable patients with full clinical follow-up. We report a chemogram sensitivity of 83.3% and specificity of 92.9%. The overall response rate and progression-free survival were higher when patients received a hit treatment as compared to patients who received a nonhit drug (as part of routine management). Finally, we leveraged our PDO collection as a platform for drug validation and combo identification. We tested anti-KRASG12D (MRTX1133), alone or combined, and identified a specific synergy with anti-EGFR therapies in KRASG12D variants. CONCLUSIONS We report the largest prospective study aiming at implementing PDO-based functional precision oncology and identify very robust predictive values in this clinical setting. In a clinically relevant turnaround time, we identify putative hits for 91% of patients, providing unexpected potential survival benefits in this very aggressive indication. Although this remains to be confirmed in interventional precision oncology trials, PDO collection already provides powerful opportunities for drugs and combinatorial treatment development.
Collapse
Affiliation(s)
- Alice Boilève
- INSERM U1279, Gustave Roussy, Villejuif, France; Université Paris Saclay, Orsay, France; Gustave Roussy, Département de Médecine, Villejuif, France.
| | - Jérôme Cartry
- INSERM U1279, Gustave Roussy, Villejuif, France; Université Paris Saclay, Orsay, France
| | - Negaar Goudarzi
- INSERM U1279, Gustave Roussy, Villejuif, France; Gustave Roussy, Plateforme Organoïdes, Villejuif, France
| | - Sabrina Bedja
- INSERM U1279, Gustave Roussy, Villejuif, France; Université Paris Saclay, Orsay, France
| | - Jacques R R Mathieu
- INSERM U1279, Gustave Roussy, Villejuif, France; Université Paris Saclay, Orsay, France
| | - Mohamed-Amine Bani
- Gustave Roussy, Département de Pathologie Morphologique, Villejuif, France
| | - Rémy Nicolle
- Centre de Recherche sur l'Inflammation, INSERM Unité 1149, Centre National de la Recherche Scientifique (CNRS), Equipe de Recherche Labellisée (ERL) 8252, Université Paris Cité, Paris, France
| | - Ali Mouawia
- INSERM U1279, Gustave Roussy, Villejuif, France
| | - Ryme Bouyakoub
- Gustave Roussy, Plateforme Organoïdes, Villejuif, France
| | - Claudio Nicotra
- Gustave Roussy, Département d'Innovations Thérapeutiques et d'Essais Précoces (DITEP), Villejuif, France
| | - Maud Ngo-Camus
- Gustave Roussy, Département d'Innovations Thérapeutiques et d'Essais Précoces (DITEP), Villejuif, France
| | - Bastien Job
- Gustave Roussy, Département de Bioinformatique, Villejuif, France
| | - Karélia Lipson
- Gustave Roussy, Plateforme Organoïdes, Villejuif, France
| | - Valérie Boige
- Gustave Roussy, Département de Médecine, Villejuif, France
| | - Marine Valéry
- Gustave Roussy, Département de Médecine, Villejuif, France
| | | | - Peggy Dartigues
- Gustave Roussy, Département de Pathologie Morphologique, Villejuif, France
| | - Lambros Tselikas
- Gustave Roussy, Département de Radiologie Interventionnelle, Villejuif, France
| | - Thierry de Baere
- Gustave Roussy, Département de Radiologie Interventionnelle, Villejuif, France
| | - Antoine Italiano
- Gustave Roussy, Département d'Innovations Thérapeutiques et d'Essais Précoces (DITEP), Villejuif, France
| | | | | | | | | | - David Malka
- INSERM U1279, Gustave Roussy, Villejuif, France; Gustave Roussy, Département de Médecine, Villejuif, France; Institut mutualiste Montsouris, Département d'Oncologie Médicale, Paris, France
| | - Cristina Smolenschi
- Gustave Roussy, Département de Médecine, Villejuif, France; Gustave Roussy, Département d'Innovations Thérapeutiques et d'Essais Précoces (DITEP), Villejuif, France
| | - Michel Ducreux
- INSERM U1279, Gustave Roussy, Villejuif, France; Université Paris Saclay, Orsay, France; Gustave Roussy, Département de Médecine, Villejuif, France
| | - Antoine Hollebecque
- Gustave Roussy, Département de Médecine, Villejuif, France; Gustave Roussy, Département d'Innovations Thérapeutiques et d'Essais Précoces (DITEP), Villejuif, France
| | - Fanny Jaulin
- INSERM U1279, Gustave Roussy, Villejuif, France; Université Paris Saclay, Orsay, France; Gustave Roussy, Département de Recherche, Villejuif, France.
| |
Collapse
|
20
|
Xiang D, He A, Zhou R, Wang Y, Xiao X, Gong T, Kang W, Lin X, Wang X, Liu L, Chen YG, Gao S, Liu Y. Building consensus on the application of organoid-based drug sensitivity testing in cancer precision medicine and drug development. Theranostics 2024; 14:3300-3316. [PMID: 38855182 PMCID: PMC11155402 DOI: 10.7150/thno.96027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024] Open
Abstract
Patient-derived organoids (PDOs) have emerged as a promising platform for clinical and translational studies. A strong correlation exists between clinical outcomes and the use of PDOs to predict the efficacy of chemotherapy and/or radiotherapy. To standardize interpretation and enhance scientific communication in the field of cancer precision medicine, we revisit the concept of PDO-based drug sensitivity testing (DST). We present an expert consensus-driven approach for medication selection aimed at predicting patient responses. To further standardize PDO-based DST, we propose guidelines for clarification and characterization. Additionally, we identify several major challenges in clinical prediction when utilizing PDOs.
Collapse
Affiliation(s)
- Dongxi Xiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200232, PRC
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, PRC
| | - Aina He
- Department of Oncology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233 PRC
| | - Rong Zhou
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200125, PRC
- National Center of Stomatology, National Clinical Research Center for Oral Disease, Shanghai 200011, PRC
| | - Yonggang Wang
- Department of Oncology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233 PRC
| | - Xiuying Xiao
- Department of Oncology, Ren Ji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, PRC
| | - Ting Gong
- Department of Oncology, Tianjin Medical University General Hospital, Tianjin 300052, PRC
| | - Wenyan Kang
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200025, PRC
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine (Boao Research Hospital), Hainan 571434, PRC
| | - Xiaolin Lin
- Department of Oncology, Ren Ji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, PRC
| | - Xiaochen Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou, Zhejiang 310009, PRC
| | | | - Lianxin Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui 230001, PRC
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui 230001, PRC
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100190, PRC
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330047, China
| | - Shaorong Gao
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200120, PRC
- Frontier Science Center for Stem Cell Research, Tongji University, 1239 Siping Road, Shanghai 200092, PRC
- Shanghai Key Laboratory of Maternal-Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, PRC
| | - Yingbin Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200232, PRC
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, PRC
| |
Collapse
|
21
|
Fu Y, Tao J, Gu Y, Liu Y, Qiu J, Su D, Wang R, Luo W, Liu T, Zhang F, Zhang T, Zhao Y. Multiomics integration reveals NETosis heterogeneity and TLR2 as a prognostic biomarker in pancreatic cancer. NPJ Precis Oncol 2024; 8:109. [PMID: 38769374 PMCID: PMC11106236 DOI: 10.1038/s41698-024-00586-x] [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: 11/08/2023] [Accepted: 03/28/2024] [Indexed: 05/22/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant neoplasm characterized by a poor prognosis and limited therapeutic strategy. The PDAC tumor microenvironment presents a complex heterogeneity, where neutrophils emerge as the predominant constituents of the innate immune cell population. Leveraging the power of single-cell RNA-seq, spatial RNA-seq, and multi-omics approaches, we included both published datasets and our in-house patient cohorts, elucidating the inherent heterogeneity in the formation of neutrophil extracellular traps (NETs) and revealed the correlation between NETs and immune suppression. Meanwhile, we constructed a multi-omics prognostic model that suggested the patients exhibiting downregulated expression of NETs may have an unfavorable outcome. We also confirmed TLR2 as a potent prognosis factor and patients with low TLR2 expression had more effective T cells and an overall survival extension for 6 months. Targeting TLR2 might be a promising strategy to reverse immunosuppression and control tumor progression for an improved prognosis.
Collapse
Affiliation(s)
- Yifan Fu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- 4 + 4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jinxin Tao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yani Gu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Beijing, 100005, China
| | - Yueze Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jiangdong Qiu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Dan Su
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ruobing Wang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Wenhao Luo
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Tao Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Feifan Zhang
- Department of Computer Science, University College London, London, UK
| | - Taiping Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
22
|
Chen HD, Ye Z, Hu HF, Fan GX, Hu YH, Li Z, Li BR, Ji SR, Zhou CJ, Xu XW, Yu XJ, Qin Y. SMAD4 endows TGF-β1-induced highly invasive tumor cells with ferroptosis vulnerability in pancreatic cancer. Acta Pharmacol Sin 2024; 45:844-856. [PMID: 38057506 PMCID: PMC10943101 DOI: 10.1038/s41401-023-01199-z] [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: 07/25/2023] [Accepted: 11/12/2023] [Indexed: 12/08/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive malignancy prone to recurrence and metastasis. Studies show that tumor cells with increased invasive and metastatic potential are more likely to undergo ferroptosis. SMAD4 is a critical molecule in the transforming growth factor β (TGF-β) pathway, which affects the TGF-β-induced epithelial-mesenchymal transition (EMT) status. SMAD4 loss is observed in more than half of patients with PDAC. In this study, we investigated whether SMAD4-positive PDAC cells were prone to ferroptosis because of their high invasiveness. We showed that SMAD4 status almost determined the orientation of transforming growth factor β1 (TGF-β1)-induced EMT via the SMAD4-dependent canonical pathway in PDAC, which altered ferroptosis vulnerability. We identified glutathione peroxidase 4 (GPX4), which inhibited ferroptosis, as a SMAD4 down-regulated gene by RNA sequencing. We found that SMAD4 bound to the promoter of GPX4 and decreased GPX4 transcription in PDAC. Furthermore, TGF-β1-induced high invasiveness enhanced sensitivity of SMAD4-positive organoids and pancreas xenograft models to the ferroptosis inducer RAS-selective lethal 3 (RSL3). Moreover, SMAD4 enhanced the cytotoxic effect of gemcitabine combined with RSL3 in highly invasive PDAC cells. This study provides new ideas for the treatment of PDAC, especially SMAD4-positive PDAC.
Collapse
Affiliation(s)
- Hai-di Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Hai-Feng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Gui-Xiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Yu-Heng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zheng Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Bo-Rui Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Shun-Rong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chen-Jie Zhou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xiao-Wu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xian-Jun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
23
|
Chi H, Su L, Yan Y, Gu X, Su K, Li H, Yu L, Liu J, Wang J, Wu Q, Yang G. Illuminating the immunological landscape: mitochondrial gene defects in pancreatic cancer through a multiomics lens. Front Immunol 2024; 15:1375143. [PMID: 38510247 PMCID: PMC10953916 DOI: 10.3389/fimmu.2024.1375143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/22/2024] Open
Abstract
This comprehensive review delves into the complex interplay between mitochondrial gene defects and pancreatic cancer pathogenesis through a multiomics approach. By amalgamating data from genomic, transcriptomic, proteomic, and metabolomic studies, we dissected the mechanisms by which mitochondrial genetic variations dictate cancer progression. Emphasis has been placed on the roles of these genes in altering cellular metabolic processes, signal transduction pathways, and immune system interactions. We further explored how these findings could refine therapeutic interventions, with a particular focus on precision medicine applications. This analysis not only fills pivotal knowledge gaps about mitochondrial anomalies in pancreatic cancer but also paves the way for future investigations into personalized therapy options. This finding underscores the crucial nexus between mitochondrial genetics and oncological immunology, opening new avenues for targeted cancer treatment strategies.
Collapse
Affiliation(s)
- Hao Chi
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Lanqian Su
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Yalan Yan
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Xiang Gu
- Biology Department, Southern Methodist University, Dallas, TX, United States
| | - Ke Su
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han Li
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Lili Yu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Jue Wang
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Qibiao Wu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Guanhu Yang
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| |
Collapse
|
24
|
Salu P, Reindl KM. Advancements in Preclinical Models of Pancreatic Cancer. Pancreas 2024; 53:e205-e220. [PMID: 38206758 PMCID: PMC10842038 DOI: 10.1097/mpa.0000000000002277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
ABSTRACT Pancreatic cancer remains one of the deadliest of all cancer types with a 5-year overall survival rate of just 12%. Preclinical models available for understanding the disease pathophysiology have evolved significantly in recent years. Traditionally, commercially available 2-dimensional cell lines were developed to investigate mechanisms underlying tumorigenesis, metastasis, and drug resistance. However, these cells grow as monolayer cultures that lack heterogeneity and do not effectively represent tumor biology. Developing patient-derived xenografts and genetically engineered mouse models led to increased cellular heterogeneity, molecular diversity, and tissues that histologically represent the original patient tumors. However, these models are relatively expensive and very timing consuming. More recently, the advancement of fast and inexpensive in vitro models that better mimic disease conditions in vivo are on the rise. Three-dimensional cultures like organoids and spheroids have gained popularity and are considered to recapitulate complex disease characteristics. In addition, computational genomics, transcriptomics, and metabolomic models are being developed to simulate pancreatic cancer progression and predict better treatment strategies. Herein, we review the challenges associated with pancreatic cancer research and available analytical models. We suggest that an integrated approach toward using these models may allow for developing new strategies for pancreatic cancer precision medicine.
Collapse
Affiliation(s)
- Philip Salu
- From the Department of Biological Sciences, North Dakota State University, Fargo, ND
| | | |
Collapse
|
25
|
Lv J, Du X, Wang M, Su J, Wei Y, Xu C. Construction of tumor organoids and their application to cancer research and therapy. Theranostics 2024; 14:1101-1125. [PMID: 38250041 PMCID: PMC10797287 DOI: 10.7150/thno.91362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
Cancer remains a severe public health burden worldwide. One of the challenges hampering effective cancer therapy is that the existing cancer models hardly recapitulate the tumor microenvironment of human patients. Over the past decade, tumor organoids have emerged as an in vitro 3D tumor model to mimic the pathophysiological characteristics of parental tumors. Various techniques have been developed to construct tumor organoids, such as matrix-based methods, hanging drop, spinner or rotating flask, nonadhesive surface, organ-on-a-chip, 3D bioprinting, and genetic engineering. This review elaborated on cell components and fabrication methods for establishing tumor organoid models. Furthermore, we discussed the application of tumor organoids to cancer modeling, basic cancer research, and anticancer therapy. Finally, we discussed current limitations and future directions in employing tumor organoids for more extensive applications.
Collapse
Affiliation(s)
- Jiajing Lv
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Institute of Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Xuan Du
- Biopharma Industry Promotion Center Shanghai, Shanghai 201203, China
| | - Miaomiao Wang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Institute of Medicine, Shanghai University, Shanghai 200444, China
- Department of Rehabilitation Medicine, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Can Xu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
26
|
Shuai Y, Zhang H, Liu C, Wang J, Jiang Y, Sun J, Gao X, Bo X, Xiao X, Liao X, Huang C, Chen H, Jiang G. CLIC3 interacts with NAT10 to inhibit N4-acetylcytidine modification of p21 mRNA and promote bladder cancer progression. Cell Death Dis 2024; 15:9. [PMID: 38182571 PMCID: PMC10770081 DOI: 10.1038/s41419-023-06373-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024]
Abstract
Chromatin accessibility plays important roles in revealing the regulatory networks of gene expression, while its application in bladder cancer is yet to be fully elucidated. Chloride intracellular channel 3 (CLIC3) protein has been reported to be associated with the progression of some tumors, whereas the specific mechanism of CLIC3 in tumor remains unclear. Here, we screened for key genes in bladder cancer through the identification of transcription factor binding site clustered region (TFCR) on the basis of chromatin accessibility and TF motif. CLIC3 was identified by joint profiling of chromatin accessibility data with TCGA database. Clinically, CLIC3 expression was significantly elevated in bladder cancer and was negatively correlated with patient survival. CLIC3 promoted the proliferation of bladder cancer cells by reducing p21 expression in vitro and in vivo. Mechanistically, CLIC3 interacted with NAT10 and inhibited the function of NAT10, resulting in the downregulation of ac4C modification and stability of p21 mRNA. Overall, these findings uncover an novel mechanism of mRNA ac4C modification and CLIC3 may act as a potential therapeutic target for bladder cancer.
Collapse
Affiliation(s)
- Yujun Shuai
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Changhao Liu
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Junting Wang
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Yangkai Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiayin Sun
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xincheng Gao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaochen Bo
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Xingyuan Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xin Liao
- Department of General Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chao Huang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Hebing Chen
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.
| | - Guosong Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
27
|
Wang H, Ning X, Zhao F, Zhao H, Li D. Human organoids-on-chips for biomedical research and applications. Theranostics 2024; 14:788-818. [PMID: 38169573 PMCID: PMC10758054 DOI: 10.7150/thno.90492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/09/2023] [Indexed: 01/05/2024] Open
Abstract
Human organoids-on-chips (OrgOCs) are the synergism of human organoids (HOs) technology and microfluidic organs-on-chips (OOCs). OOCs can mimic extrinsic characteristics of organs, such as environmental clues of living tissue, while HOs are more amenable to biological analysis and genetic manipulation. By spatial cooperation, OrgOCs served as 3D organotypic living models allowing them to recapitulate critical tissue-specific properties and forecast human responses and outcomes. It represents a giant leap forward from the regular 2D cell monolayers and animal models in the improved human ecological niche modeling. In recent years, OrgOCs have offered potential promises for clinical studies and advanced the preclinical-to-clinical translation in medical and industrial fields. In this review, we highlight the cutting-edge achievements in OrgOCs, introduce the key features of OrgOCs architectures, and share the revolutionary applications in basic biology, disease modeling, preclinical assay and precision medicine. Furthermore, we discuss how to combine a wide range of disciplines with OrgOCs and accelerate translational applications, as well as the challenges and opportunities of OrgOCs in biomedical research and applications.
Collapse
Affiliation(s)
- Hui Wang
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Xiufan Ning
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Feng Zhao
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Hui Zhao
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Dong Li
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| |
Collapse
|
28
|
Song T, Kong B, Liu R, Luo Y, Wang Y, Zhao Y. Bioengineering Approaches for the Pancreatic Tumor Organoids Research and Application. Adv Healthc Mater 2024; 13:e2300984. [PMID: 37694339 DOI: 10.1002/adhm.202300984] [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: 04/26/2023] [Revised: 08/29/2023] [Indexed: 09/12/2023]
Abstract
Pancreatic cancer is a highly lethal form of digestive malignancy that poses significant health risks to individuals worldwide. Chemotherapy-based comprehensive treatment is the primary therapeutic approach for midlife and late-life patients. Nevertheless, the heterogeneity of the tumor and individual genetic backgrounds result in substantial variations in drug sensitivity among patients, rendering a single treatment regimen unsuitable for all patients. Conventional pancreatic cancer tumor organoid models are capable of emulating the biological traits of pancreatic cancer and are utilized in drug development and screening. However, these tumor organoids can still not mimic the tumor microenvironment (TME) in vivo, and the poor controllability in the preparation process hinders translation from essential drug screening to clinical pharmacological therapy. In recent years, many engineering methods with remarkable results have been used to develop pancreatic cancer organoid models, including bio-hydrogel, co-culture, microfluidic, and gene editing. Here, this work summarizes and analyzes the recent developments in engineering pancreatic tumor organoid models. In addition, the future direction of improving engineered pancreatic cancer organoids is discussed for their application prospects in clinical treatment.
Collapse
Affiliation(s)
- Taiyu Song
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Bin Kong
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Rui Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
29
|
Gómez-Álvarez M, Agustina-Hernández M, Francés-Herrero E, Rodríguez-Eguren A, Bueno-Fernandez C, Cervelló I. Addressing Key Questions in Organoid Models: Who, Where, How, and Why? Int J Mol Sci 2023; 24:16014. [PMID: 37958996 PMCID: PMC10650475 DOI: 10.3390/ijms242116014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Organoids are three-dimensional cellular structures designed to recreate the biological characteristics of the body's native tissues and organs in vitro. There has been a recent surge in studies utilizing organoids due to their distinct advantages over traditional two-dimensional in vitro approaches. However, there is no consensus on how to define organoids. This literature review aims to clarify the concept of organoids and address the four fundamental questions pertaining to organoid models: (i) What constitutes organoids?-The cellular material. (ii) Where do organoids grow?-The extracellular scaffold. (iii) How are organoids maintained in vitro?-Via the culture media. (iv) Why are organoids suitable in vitro models?-They represent reproducible, stable, and scalable models for biological applications. Finally, this review provides an update on the organoid models employed within the female reproductive tract, underscoring their relevance in both basic biology and clinical applications.
Collapse
Affiliation(s)
- María Gómez-Álvarez
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
| | - Marcos Agustina-Hernández
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
| | - Emilio Francés-Herrero
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
- Department of Pediatrics, Obstetrics and Gynecology, Universitat de València, 46010 Valencia, Spain
| | - Adolfo Rodríguez-Eguren
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
| | - Clara Bueno-Fernandez
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
- Department of Pediatrics, Obstetrics and Gynecology, Universitat de València, 46010 Valencia, Spain
| | - Irene Cervelló
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
| |
Collapse
|
30
|
Hao X, Zu M, Ning J, Zhou X, Gong Y, Han X, Meng Q, Li D, Ding S. Antitumor effect of luteolin proven by patient-derived organoids of gastric cancer. Phytother Res 2023; 37:5315-5327. [PMID: 37469042 DOI: 10.1002/ptr.7963] [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: 03/06/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/21/2023]
Abstract
Luteolin (Lut) has been shown to inhibit gastric cancer (GC); however, its efficacy compared to other clinical drugs has not been examined in human samples. This study aimed to elucidate the antitumor activity of Lut in GC patient-derived organoids (PDOs). PDOs were established from GC cancer tissues, and the characterization of tissues and PDOs was performed using whole-exome sequencing. Drug sensitivity tests were performed by treating PDOs with Lut, norcantharidin (NCTD), and carboplatin (CP). RNA sequencing of PDOs was performed to elucidate the antitumor mechanism of Lut, which was further verified in three GC cell lines. Eleven PDOs were successfully constructed, and were highly consistent with the pathophysiology and genetic changes in the corresponding tumors. The IC50s of Lut, NCTD, and CP of PDOs were 27.19, 23.9, and 37.87 μM, respectively. Lut treatment upregulated FOXO3, DUSP1, and CDKN1A expression and downregulated IL1R1 and FGFR4 expression in GC cell lines, which was consistent with the results of PDOs. We demonstrate that Lut exerted stronger antitumor effects than CP, but a similar effect to that of NCTD, which was obtained in an in vitro PDO system. Additionally, Lut exerted varying degrees of antitumor effects against the PDOs, thereby indicating that PDO may be a useful preclinical drug screening tool for personalized treatment.
Collapse
Affiliation(s)
- Xinyu Hao
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| | - Ming Zu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| | - Jing Ning
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| | - Xin Zhou
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Yueqing Gong
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| | - Xiurui Han
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| | - Qiao Meng
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| | - Dong Li
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Shigang Ding
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| |
Collapse
|
31
|
Jiang W, Gu G, Zhang Y, Song Y, Shi M, Wang G, Li H, Tao T, Qin J, Li X, Jia H, Jiao F, Xu W, Huang X. Novel mesothelin-targeted chimeric antigen receptor-modified UNKT cells are highly effective in inhibiting tumor progression. Pharmacol Res 2023; 197:106942. [PMID: 37775021 DOI: 10.1016/j.phrs.2023.106942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/01/2023]
Abstract
The design of chimeric antigen receptors (CAR) significantly enhances the antitumor efficacy of T cells. Although some CAR-T products have been approved by FDA in treating hematological tumors, adoptive immune therapy still faces many difficulties and challenges in the treatment of solid tumors. In this study, we reported a new strategy to treat solid tumors using a natural killer-like T (NKT) cell line which showed strong cytotoxicity to lyse 15 cancer cell lines, safe to normal cells and had low or no Graft-versus-host activity. We thus named it as universal NKT (UNKT). In both direct and indirect 3D tumor-like organ model, UNKT showed efficient tumor-killing properties, indicating that it could penetrate the microenvironment of solid tumors. In mesothelin (MSLN)-positive tumor cells (SKOV-3 and MCF-7), MSLN targeting CAR modified-UNKT cells had enhanced killing potential against MSLN positive ovarian cancer compared with the wild type UNKT, as well as MSLN-CAR-T cells. Compared with CAR-T, Single-cell microarray 32-plex proteomics revealed CAR-UNKT cells express more effector cytokines, such as perforin and granzyme B, and less interleukin-6 after activation. Moreover, our CAR-UNKT cells featured in more multifunctionality than CAR-T cells. CAR-UNKT cells also demonstrated strong antitumor activity in mouse models of ovarian cancer, with the ability to migrate and infiltrate the tumor without inducing immune memory. The fast-in and -out, enhanced and prolonged tumor killing properties of CAR-UNKT suggested a novel cure option of cellular immunotherapy in the treatment of MSLN-positive solid tumors.
Collapse
Affiliation(s)
- Wei Jiang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Guosheng Gu
- Abelow Pharmaceuticals Inc., 10 Xinghuo Road, Jiangbei New Area, Nanjing, Jiangsu 210000, China
| | - Yumin Zhang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Yushuai Song
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Ming Shi
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Tingting Tao
- CAS Key Laboratory of SSAC, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Jianhua Qin
- CAS Key Laboratory of SSAC, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; Beijing Institute For Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100020, China; Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215000, China; University of Chinese Academy of Sciences, Beijing 100020, China
| | - Xianliang Li
- Department of HBP Surgery,Beijing Chao Yang Hospital,The Capital Medical University, Beijing 100020, China
| | - Hongtao Jia
- Abelow Pharmaceuticals Inc., 10 Xinghuo Road, Jiangbei New Area, Nanjing, Jiangsu 210000, China
| | - Feng Jiao
- Abelow Pharmaceuticals Inc., 10 Xinghuo Road, Jiangbei New Area, Nanjing, Jiangsu 210000, China
| | - Weidong Xu
- Abelow Pharmaceuticals Inc., 10 Xinghuo Road, Jiangbei New Area, Nanjing, Jiangsu 210000, China.
| | - Xiaoyi Huang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin 150081, China; NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China.
| |
Collapse
|
32
|
Sun L, Kang X, Ju H, Wang C, Yang G, Wang R, Sun S. A human mucosal melanoma organoid platform for modeling tumor heterogeneity and exploring immunotherapy combination options. SCIENCE ADVANCES 2023; 9:eadg6686. [PMID: 37889972 PMCID: PMC10610903 DOI: 10.1126/sciadv.adg6686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023]
Abstract
Mucosal melanoma (MM), an aggressive rare subtype of melanoma, is distinct from cutaneous melanoma and has poor prognoses. We addressed the lack of cell models for MM by establishing 30 organoids of human oral MM (OMM), which retained major histopathological and functional features of parental tumors. Organoid groups derived from chronologically or intratumorally distinct lesions within the same individual displayed heterogeneous genetics, expression profiles, and drug responses, indicating rapid tumor evolution and poor clinical response. Furthermore, transcriptome analysis revealed receptor tyrosine kinases (RTKs) signaling, particularly NGFR, a nerve growth factor receptor, was significantly up-regulated in OMMs and organoids from patients resistant to anti-programmed cell death protein 1 (anti-PD-1) therapy. Combining anti-PD-1 with anlotinib (a phase 2 multitarget RTK inhibitor for OMM) or NGFR knockdown enhanced the effective activity of immune cells in organoid-immune cell coculture systems. Together, our study suggested that OMM organoids serve as faithful models for exploring tumor evolution and immunotherapy combination strategies.
Collapse
Affiliation(s)
- Lulu Sun
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
- Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Xindan Kang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
- Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Houyu Ju
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
- Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Chong Wang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
- Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Guizhu Yang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
- Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Rui Wang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
- Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Shuyang Sun
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
- Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| |
Collapse
|
33
|
Gong M, Meng H, Tan D, Li P, Qin J, An Q, Shi C, An J. Establishment of organoid models for pancreatic ductal adenocarcinoma and screening of individualized therapy strategy. Animal Model Exp Med 2023; 6:409-418. [PMID: 37890865 PMCID: PMC10614126 DOI: 10.1002/ame2.12352] [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: 06/01/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Patients with pancreatic ductal adenocarcinoma (PDAC) who undergo surgical resection and receive effective chemotherapy have the best chance for long-term survival. Unfortunately, because of the heterogeneity of pancreatic cancer, it is difficult to find a personalized treatment strategy for patients. Organoids are ideal preclinical models for personalized medicine. Therefore, we explore the cultivation conditions and construction methods of PDAC organoid models to screen the individualized therapy strategy. METHODS Fresh PDAC tissues from surgical resection were collected and digested with digestive enzymes; then the tumor cells were embedded in Matrigel with a suitable medium to establish the PDAC organoid models. The genetic stability of the organoids was analyzed using whole exon sequencing; hematoxylin and eosin staining and immunohistochemistry of organoids were performed to analyze their consistency with the pathological morphology of the patient's tumor tissue; After 2 days of organoid culture, we selected four commonly used clinical chemotherapy drugs for single or combined treatment to analyze drug sensitivity. RESULTS Two cases of PDAC organoid models were successfully established, and the results of their pathological characteristics and exome sequencing were consistent with those of the patient's tumor tissue. Both PDAC organoids showed more sensitivity to gemcitabine and cisplatin, and the combined treatment was more effective than monotherapy. CONCLUSION Both organoids better retained the pathological characteristics, genomic stability, and heterogeneity with the original tumor. Individual PDAC organoids exhibited different sensitivities to the same drugs. Thus, this study provided ideal experimental models for screening individualized therapy strategy for patients with PDAC.
Collapse
Affiliation(s)
- Miaomiao Gong
- Division of Cancer Biology, Laboratory Animal CenterFourth Military Medical UniversityXi'anChina
- School of Basic Medical SciencesMedical College of Yan'an UniversityYananChina
| | - Han Meng
- Division of Cancer Biology, Laboratory Animal CenterFourth Military Medical UniversityXi'anChina
| | - Dengxu Tan
- Division of Cancer Biology, Laboratory Animal CenterFourth Military Medical UniversityXi'anChina
| | - Peng Li
- Division of Cancer Biology, Laboratory Animal CenterFourth Military Medical UniversityXi'anChina
- Animal Experiment CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Jing Qin
- Division of Cancer Biology, Laboratory Animal CenterFourth Military Medical UniversityXi'anChina
| | - Qingling An
- Division of Cancer Biology, Laboratory Animal CenterFourth Military Medical UniversityXi'anChina
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal CenterFourth Military Medical UniversityXi'anChina
| | - Jiaze An
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Xijing HospitalFourth Military Medical UniversityXi'anChina
| |
Collapse
|
34
|
Li Y, Tang S, Shi X, Lv J, Wu X, Zhang Y, Wang H, He J, Zhu Y, Ju Y, Zhang Y, Guo S, Yang W, Yin H, Chen L, Gao D, Jin G. Metabolic classification suggests the GLUT1/ALDOB/G6PD axis as a therapeutic target in chemotherapy-resistant pancreatic cancer. Cell Rep Med 2023; 4:101162. [PMID: 37597521 PMCID: PMC10518604 DOI: 10.1016/j.xcrm.2023.101162] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 06/14/2023] [Accepted: 07/26/2023] [Indexed: 08/21/2023]
Abstract
Metabolic reprogramming is known as an emerging mechanism of chemotherapy resistance, but the metabolic signatures of pancreatic ductal adenocarcinomas (PDACs) remain unclear. Here, we characterize the metabolomic profile of PDAC organoids and classify them into glucomet-PDAC (high glucose metabolism levels) and lipomet-PDAC (high lipid metabolism levels). Glucomet-PDACs are more resistant to chemotherapy than lipomet-PDACs, and patients with glucomet-PDAC have a worse prognosis. Integrated analyses reveal that the GLUT1/aldolase B (ALDOB)/glucose-6-phosphate dehydrogenase (G6PD) axis induces chemotherapy resistance by remodeling glucose metabolism in glucomet-PDAC. Increased glycolytic flux, G6PD activity, and pyrimidine biosynthesis are identified in glucomet-PDAC with high GLUT1 and low ALDOB expression, and these phenotypes could be reversed by inhibiting GLUT1 expression or by increasing ALDOB expression. Pharmacological inhibition of GLUT1 or G6PD enhances the chemotherapy response of glucomet-PDAC. Our findings uncover potential metabolic heterogeneity related to differences in chemotherapy sensitivity in PDAC and develop a promising pharmacological strategy for patients with chemotherapy-resistant glucomet-PDAC through the combination of chemotherapy and GLUT1/ALDOB/G6PD axis inhibitors.
Collapse
Affiliation(s)
- Yunguang Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shijie Tang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Jingwen Lv
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety Research, Shanghai Institute of Nutrition and Health (SINH), Innovation Center for Intervention of Chronic Disease and Promotion of Health, Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Xueyuan Wu
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yehan Zhang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huan Wang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Juan He
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiqin Zhu
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yi Ju
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yajuan Zhang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shiwei Guo
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Weiwei Yang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Huiyong Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety Research, Shanghai Institute of Nutrition and Health (SINH), Innovation Center for Intervention of Chronic Disease and Promotion of Health, Chinese Academy of Sciences (CAS), Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China.
| | - Luonan Chen
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Dong Gao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China.
| |
Collapse
|
35
|
Rana M, Kansal RG, Bisunke B, Fang J, Shibata D, Bajwa A, Yang J, Glazer ES. Bromo- and Extra-Terminal Domain Inhibitors Induce Mitochondrial Stress in Pancreatic Ductal Adenocarcinoma. Mol Cancer Ther 2023; 22:936-946. [PMID: 37294884 PMCID: PMC10527726 DOI: 10.1158/1535-7163.mct-23-0149] [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: 03/09/2023] [Revised: 04/28/2023] [Accepted: 06/06/2023] [Indexed: 06/11/2023]
Abstract
Identifying novel, unique, and personalized molecular targets for patients with pancreatic ductal adenocarcinoma (PDAC) remains the greatest challenge in altering the biology of fatal tumors. Bromo- and extra-terminal domain (BET) proteins are activated in a noncanonical fashion by TGFβ, a ubiquitous cytokine in the PDAC tumor microenvironment (TME). We hypothesized that BET inhibitors (BETi) represent a new class of drugs that attack PDAC tumors via a novel mechanism. Using a combination of patient and syngeneic murine models, we investigated the effects of the BETi drug BMS-986158 on cellular proliferation, organoid growth, cell-cycle progression, and mitochondrial metabolic disruption. These were investigated independently and in combination with standard cytotoxic chemotherapy (gemcitabine + paclitaxel [GemPTX]). BMS-986158 reduced cell viability and proliferation across multiple PDAC cell lines in a dose-dependent manner, even more so in combination with cytotoxic chemotherapy (P < 0.0001). We found that BMS-986158 reduced both human and murine PDAC organoid growth (P < 0.001), with associated perturbations in the cell cycle leading to cell-cycle arrest. BMS-986158 disrupts normal cancer-dependent mitochondrial function, leading to aberrant mitochondrial metabolism and stress via dysfunctional cellular respiration, proton leakage, and ATP production. We demonstrated mechanistic and functional data that BETi induces metabolic mitochondrial dysfunction, abrogating PDAC progression and proliferation, alone and in combination with systemic cytotoxic chemotherapies. This novel approach improves the therapeutic window in patients with PDAC and offers another treatment approach distinct from cytotoxic chemotherapy that targets cancer cell bioenergetics.
Collapse
Affiliation(s)
- Manjul Rana
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Rita G. Kansal
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Bijay Bisunke
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Jie Fang
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN
| | - David Shibata
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Amandeep Bajwa
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Department of Genetics, Genomics, and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Jun Yang
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN
- Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Department of Genetics, Genomics, and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Comprehensive Cancer Center, St. Jude Children’s Research Hospital, Memphis, TN
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, St. Jude Children’s Research Hospital, Memphis, TN
| | - Evan S. Glazer
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| |
Collapse
|
36
|
Forsythe SD, Pu T, Andrews SG, Madigan JP, Sadowski SM. Models in Pancreatic Neuroendocrine Neoplasms: Current Perspectives and Future Directions. Cancers (Basel) 2023; 15:3756. [PMID: 37568572 PMCID: PMC10416968 DOI: 10.3390/cancers15153756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/21/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
Pancreatic neuroendocrine neoplasms (pNENs) are a heterogeneous group of tumors derived from multiple neuroendocrine origin cell subtypes. Incidence rates for pNENs have steadily risen over the last decade, and outcomes continue to vary widely due to inability to properly screen. These tumors encompass a wide range of functional and non-functional subtypes, with their rarity and slow growth making therapeutic development difficult as most clinically used therapeutics are derived from retrospective analyses. Improved molecular understanding of these cancers has increased our knowledge of the tumor biology for pNENs. Despite these advances in our understanding of pNENs, there remains a dearth of models for further investigation. In this review, we will cover the current field of pNEN models, which include established cell lines, animal models such as mice and zebrafish, and three-dimensional (3D) cell models, and compare their uses in modeling various disease aspects. While no study model is a complete representation of pNEN biology, each has advantages which allow for new scientific understanding of these rare tumors. Future efforts and advancements in technology will continue to create new options in modeling these cancers.
Collapse
Affiliation(s)
- Steven D. Forsythe
- Neuroendocrine Cancer Therapy Section, Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (S.D.F.); (S.G.A.); (J.P.M.)
| | - Tracey Pu
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Stephen G. Andrews
- Neuroendocrine Cancer Therapy Section, Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (S.D.F.); (S.G.A.); (J.P.M.)
| | - James P. Madigan
- Neuroendocrine Cancer Therapy Section, Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (S.D.F.); (S.G.A.); (J.P.M.)
| | - Samira M. Sadowski
- Neuroendocrine Cancer Therapy Section, Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (S.D.F.); (S.G.A.); (J.P.M.)
| |
Collapse
|
37
|
Ekeuku SO, Etim EP, Pang KL, Chin KY, Mai CW. Vitamin E in the management of pancreatic cancer: A scoping review. World J Gastrointest Oncol 2023; 15:943-958. [PMID: 37389119 PMCID: PMC10302993 DOI: 10.4251/wjgo.v15.i6.943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/03/2023] [Accepted: 04/07/2023] [Indexed: 06/14/2023] Open
Abstract
Pancreatic cancer is the leading cause of cancer mortality worldwide. Research investigating effective management strategies for pancreatic cancer is ongoing. Vitamin E, consisting of both tocopherol and tocotrienol, has demonstrated debatable effects on pancreatic cancer cells. Therefore, this scoping review aims to summarize the effects of vitamin E on pancreatic cancer. In October 2022, a literature search was conducted using PubMed and Scopus since their inception. Original studies on the effects of vitamin E on pancreatic cancer, including cell cultures, animal models and human clinical trials, were considered for this review. The literature search found 75 articles on this topic, but only 24 articles met the inclusion criteria. The available evidence showed that vitamin E modulated proliferation, cell death, angiogenesis, metastasis and inflammation in pancreatic cancer cells. However, the safety and bioavailability concerns remain to be answered with more extensive preclinical and clinical studies. More in-depth analysis is necessary to investigate further the role of vitamin E in the management of pancreatic cancers.
Collapse
Affiliation(s)
- Sophia Ogechi Ekeuku
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Effiong Paul Etim
- Faculty of Applied Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Kok-Lun Pang
- Newcastle University Medicine Malaysia, Iskandar Puteri 79200, Johor, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Chun-Wai Mai
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
38
|
Huang P, Gao W, Fu C, Tian R. Functional and Clinical Proteomic Exploration of Pancreatic Cancer. Mol Cell Proteomics 2023:100575. [PMID: 37209817 PMCID: PMC10388587 DOI: 10.1016/j.mcpro.2023.100575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/18/2023] [Accepted: 05/11/2023] [Indexed: 05/22/2023] Open
Abstract
Pancreatic cancer, most cases being pancreatic ductal adenocarcinoma (PDAC), is one of the most lethal cancers with a median survival time of less than 6 months. Therapeutic options are very limited for PDAC patients, and surgery is still the most effective treatment, making improvements in early diagnosis critical. One typical characteristic of PDAC is the desmoplastic reaction of its stroma microenvironment, which actively interacts with cancer cells to orchestrate key components in tumorigenesis, metastasis, and chemoresistance. Global exploration of cancer-stroma crosstalk is essential to decipher PDAC biology and design intervention strategies. Over the past decade, the dramatic improvement of proteomics technologies has enabled profiling of proteins, post-translational modifications (PTMs), and their protein complexes at unprecedented sensitivity and dimensionality. Here, starting with our current understanding of PDAC characteristics, including precursor lesions, progression models, tumor microenvironment, and therapeutic advancements, we describe how proteomics contributes to the functional and clinical exploration of PDAC, providing insights into PDAC carcinogenesis, progression, and chemoresistance. We summarize recent achievements enabled by proteomics to systematically investigate PTMs-mediated intracellular signaling in PDAC, cancer-stroma interactions, and potential therapeutic targets revealed by these functional studies. We also highlight proteomic profiling of clinical tissue and plasma samples to discover and verify useful biomarkers that can aid early detection and molecular classification of patients. In addition, we introduce spatial proteomic technology and its applications in PDAC for deconvolving tumor heterogeneity. Finally, we discuss future prospects of applying new proteomic technologies in comprehensively understanding PDAC heterogeneity and intercellular signaling networks. Importantly, we expect advances in clinical functional proteomics for exploring mechanisms of cancer biology directly by high-sensitivity functional proteomic approaches starting from clinical samples.
Collapse
Affiliation(s)
- Peiwu Huang
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Weina Gao
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Changying Fu
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ruijun Tian
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
39
|
Azim R, Wang S, Dipu SA, Islam N, Ala Muid MR, Elahe MF. A patient-specific functional module and path identification technique from RNA-seq data. Comput Biol Med 2023; 158:106871. [PMID: 37030265 DOI: 10.1016/j.compbiomed.2023.106871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/12/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023]
Abstract
With the advancement of new technologies, a huge amount of high dimensional data is being generated which is opening new opportunities and challenges to the study of cancer and diseases. In particular, distinguishing the patient-specific key components and modules which drive tumorigenesis is necessary to analyze. A complex disease generally does not initiate from the dysregulation of a single component but it is the result of the dysfunction of a group of components and networks which differs from patient to patient. However, a patient-specific network is required to understand the disease and its molecular mechanism. We address this requirement by constructing a patient-specific network by sample-specific network theory with integrating cancer-specific differentially expressed genes and elite genes. By elucidating patient-specific networks, it can identify the regulatory modules, driver genes as well as personalized disease networks which can lead to personalized drug design. This method can provide insight into how genes are associating with each other and characterized the patient-specific disease subtypes. The results show that this method can be beneficial for the detection of patient-specific differential modules and interaction between genes. Extensive analysis using existing literature, gene enrichment and survival analysis for three cancer types STAD, PAAD and LUAD shows the effectiveness of this method over other existing methods. In addition, this method can be useful for personalized therapeutics and drug design. This methodology is implemented in the R language and is available at https://github.com/riasatazim/PatientSpecificRNANetwork.
Collapse
|
40
|
Lin DC. Large-scale genomic analyses reveal alterations and mechanisms underlying clonal evolution and immune evasion in esophageal cancer. Nat Commun 2023; 14:893. [PMID: 36807265 PMCID: PMC9938131 DOI: 10.1038/s41467-023-36557-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/23/2022] [Indexed: 02/19/2023] Open
Affiliation(s)
- De-Chen Lin
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA.
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Liu Y, Li N, Zhu Y. Pancreatic Organoids: A Frontier Method for Investigating Pancreatic-Related Diseases. Int J Mol Sci 2023; 24:4027. [PMID: 36835437 PMCID: PMC9959977 DOI: 10.3390/ijms24044027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
The pancreas represents an important organ that has not been comprehensively studied in many fields. To fill this gap, many models have been generated, and traditional models have shown good performance in addressing pancreatic-related diseases, but are increasingly struggling to keep up with the need for further research due to ethical issues, genetic heterogeneity and difficult clinical translation. The new era calls for new and more reliable research models. Therefore, organoids have been proposed as a novel model for the evaluation of pancreatic-related diseases such as pancreatic malignancy, diabetes, and pancreatic cystic fibrosis. Compared with common traditional models, including 2D cell culture and gene editing mice, organoids derived from living humans or mice cause minimal harm to the donor, raise fewer ethical concerns, and reasonably address the claims of heterogeneity, which allows for the further development of pathogenesis studies and clinical trial analysis. In this review, we analyse studies on the use of pancreatic organoids in research on pancreatic-related diseases, discuss the advantages and disadvantages, and hypothesize future trends.
Collapse
Affiliation(s)
- Yuxiang Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330209, China
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang 330209, China
| | - Nianshuang Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330209, China
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang 330209, China
- Jiangxi Institute of Digestive Disease, The First Affiliated Hospital of Nanchang University, Nanchang 330209, China
| | - Yin Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang 330209, China
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang 330209, China
| |
Collapse
|
42
|
Wang Y, Wang F, Qin Y, Lou X, Ye Z, Zhang W, Gao H, Chen J, Xu X, Yu X, Ji S. Recent progress of experimental model in pancreatic neuroendocrine tumors: drawbacks and challenges. Endocrine 2023; 80:266-282. [PMID: 36648608 DOI: 10.1007/s12020-023-03299-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/31/2022] [Indexed: 01/18/2023]
Abstract
The neuroendocrine neoplasm, in general, refers to a heterogeneous group of all tumors originating from peptidergic neurons and neuroendocrine cells. Neuroendocrine neoplasms are divided into two histopathological subtypes: well-differentiated neuroendocrine tumors and poorly differentiated neuroendocrine carcinomas. Pancreatic neuroendocrine tumors account for more than 80% of pancreatic neuroendocrine neoplasms. Due to the greater proportion of pancreatic neuroendocrine tumors compared to pancreatic neuroendocrine carcinoma, this review will only focus on them. The worldwide incidence of pancreatic neuroendocrine tumors is rising year by year due to sensitive detection with an emphasis on medical examinations and the improvement of testing technology. Although the biological behavior of pancreatic neuroendocrine tumors tends to be inert, distant metastasis is common, often occurring very early. Because of the paucity of basic research on pancreatic neuroendocrine tumors, the mechanism of tumor development, metastasis, and recurrence are still unclear. In this context, the representative preclinical models simulating the tumor development process are becoming ever more widely appreciated to address the clinical problems of pancreatic neuroendocrine tumors. So far, there is no comprehensive report on the experimental model of pancreatic neuroendocrine tumors. This article systematically summarizes the characteristics of preclinical models, such as patient-derived cell lines, patient-derived xenografts, genetically engineered mouse models, and patient-derived organoids, and their advantages and disadvantages, to provide a reference for further studies of neuroendocrine tumors. We also highlight the method of establishment of liver metastasis mouse models.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Fei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xin Lou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Wuhu Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Heli Gao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jie Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
43
|
Yu C, Kang R, Tang D. Organoids Models of Pancreatic Duct Adenocarcinoma. Methods Mol Biol 2023; 2712:45-60. [PMID: 37578695 DOI: 10.1007/978-1-0716-3433-2_5] [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: 08/15/2023]
Abstract
Three-dimensional (3D) organoid culture is a laboratory technique used to grow and study miniature organs that mimic the structure and function of real organs in the human body. Organoids are created from stem cells or tissue samples and are grown in a 3D matrix that allows them to self-organize into a complex, three-dimensional structure. Organoids are valuable tools for studying human biology and disease, including cancer. Pancreatic ductal adenocarcinoma (PDAC) still has the worst survival rate of common malignancies, despite recent advances in cancer treatment. Preclinical studies have shown that impaired cell death pathways, including apoptosis, necroptosis, ferroptosis, pyroptosis, and alkaliptosis, promote PDAC development. Organoid models are now widely used in the study of pancreatic cancer biology, including cell death machinery. This chapter provides step-by-step protocols for generating human or mice PDAC organoids in a 3D Matrigel system.
Collapse
Affiliation(s)
- Chunhua Yu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
44
|
Raut P, Nimmakayala RK, Batra SK, Ponnusamy MP. Clinical and Molecular Attributes and Evaluation of Pancreatic Cystic Neoplasm. Biochim Biophys Acta Rev Cancer 2023; 1878:188851. [PMID: 36535512 PMCID: PMC9898173 DOI: 10.1016/j.bbcan.2022.188851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/08/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Intraductal papillary mucinous neoplasms (IPMNs) and mucinous cystic neoplasms (MCNs) are all considered "Pancreatic cystic neoplasms (PCNs)" and show a varying risk of developing into pancreatic ductal adenocarcinoma (PDAC). These lesions display different molecular characteristics, mutations, and clinical manifestations. A lack of detailed understanding of PCN subtype characteristics and their molecular mechanisms limits the development of efficient diagnostic tools and therapeutic strategies for these lesions. Proper in vivo mouse models that mimic human PCNs are also needed to study the molecular mechanisms and for therapeutic testing. A comprehensive understanding of the current status of PCN biology, mechanisms, current diagnostic methods, and therapies will help in the early detection and proper management of patients with these lesions and PDAC. This review aims to describe all these aspects of PCNs, specifically IPMNs, by describing the future perspectives.
Collapse
Affiliation(s)
- Pratima Raut
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| |
Collapse
|
45
|
Nam C, Ziman B, Sheth M, Zhao H, Lin DC. Genomic and Epigenomic Characterization of Tumor Organoid Models. Cancers (Basel) 2022; 14:4090. [PMID: 36077628 PMCID: PMC9454968 DOI: 10.3390/cancers14174090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Tumor organoid modeling has been recognized as a state-of-the-art system for in vitro research on cancer biology and precision oncology. Organoid culture technologies offer distinctive advantages, including faithful maintenance of physiological and pathological characteristics of human disease, self-organization into three-dimensional multicellular structures, and preservation of genomic and epigenomic landscapes of the originating tumor. These features effectively position organoid modeling between traditional cell line cultures in two dimensions and in vivo animal models as a valid, versatile, and robust system for cancer research. Here, we review recent advances in genomic and epigenomic characterization of tumor organoids and the novel findings obtained, highlight significant progressions achieved in organoid modeling of gene-drug interactions and genotype-phenotype associations, and offer perspectives on future opportunities for organoid modeling in basic and clinical cancer research.
Collapse
Affiliation(s)
| | | | | | | | - De-Chen Lin
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|