1
|
Gu A, Li J, Li M, Liu Y. Patient-derived xenograft model in cancer: establishment and applications. MedComm (Beijing) 2025; 6:e70059. [PMID: 39830019 PMCID: PMC11742426 DOI: 10.1002/mco2.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/24/2024] [Accepted: 12/15/2024] [Indexed: 01/22/2025] Open
Abstract
The patient-derived xenograft (PDX) model is a crucial in vivo model extensively employed in cancer research that has been shown to maintain the genomic characteristics and pathological structure of patients across various subtypes, metastatic, and diverse treatment histories. Various treatment strategies utilized in PDX models can offer valuable insights into the mechanisms of tumor progression, drug resistance, and the development of novel therapies. This review provides a comprehensive overview of the establishment and applications of PDX models. We present an overview of the history and current status of PDX models, elucidate the diverse construction methodologies employed for different tumors, and conduct a comparative analysis to highlight the distinct advantages and limitations of this model in relation to other in vivo models. The applications are elucidated in the domain of comprehending the mechanisms underlying tumor development and cancer therapy, which highlights broad applications in the fields of chemotherapy, targeted therapy, delivery systems, combination therapy, antibody-drug conjugates and radiotherapy. Furthermore, the combination of the PDX model with multiomics and single-cell analyses for cancer research has also been emphasized. The application of the PDX model in clinical treatment and personalized medicine is additionally emphasized.
Collapse
Affiliation(s)
- Ao Gu
- Department of Biliary‐Pancreatic SurgeryRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jiatong Li
- Department of Biliary‐Pancreatic SurgeryRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Meng‐Yao Li
- Department of Biliary‐Pancreatic SurgeryRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yingbin Liu
- Department of Biliary‐Pancreatic SurgeryRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
2
|
Janic A, Abad E, Amelio I. Decoding p53 tumor suppression: a crosstalk between genomic stability and epigenetic control? Cell Death Differ 2025; 32:1-8. [PMID: 38379088 PMCID: PMC11742645 DOI: 10.1038/s41418-024-01259-9] [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: 10/03/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
Genomic instability, a hallmark of cancer, is a direct consequence of the inactivation of the tumor suppressor protein p53. Genetically modified mouse models and human tumor samples have revealed that p53 loss results in extensive chromosomal abnormalities, from copy number alterations to structural rearrangements. In this perspective article we explore the multifaceted relationship between p53, genomic stability, and epigenetic control, highlighting its significance in cancer biology. p53 emerges as a critical regulator of DNA repair mechanisms, influencing key components of repair pathways and directly participating in DNA repair processes. p53 role in genomic integrity however extends beyond its canonical functions. p53 influences also epigenetic landscape, where it modulates DNA methylation and histone modifications. This epigenetic control impacts the expression of genes involved in tumor suppression and oncogenesis. Notably, p53 ability to ensure cellular response to DNA demethylation contributes to the maintenance of genomic stability by preventing unscheduled transcription of repetitive non-coding genomic regions. This latter indicates a causative relationship between the control of epigenetic stability and the maintenance of genomic integrity in p53-mediated tumor suppression. Understanding these mechanisms offers promising avenues for innovative therapeutic strategies targeting epigenetic dysregulation in cancer and emphasizes the need for further research to unravel the complexities of this relationship. Ultimately, these insights hold the potential to transform cancer treatment and prevention strategies.
Collapse
Affiliation(s)
- Ana Janic
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
| | - Etna Abad
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ivano Amelio
- Chair for Systems Toxicology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
3
|
Wang H, Li X, You X, Zhao G. Harnessing the power of artificial intelligence for human living organoid research. Bioact Mater 2024; 42:140-164. [PMID: 39280585 PMCID: PMC11402070 DOI: 10.1016/j.bioactmat.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/21/2024] [Accepted: 08/26/2024] [Indexed: 09/18/2024] Open
Abstract
As a powerful paradigm, artificial intelligence (AI) is rapidly impacting every aspect of our day-to-day life and scientific research through interdisciplinary transformations. Living human organoids (LOs) have a great potential for in vitro reshaping many aspects of in vivo true human organs, including organ development, disease occurrence, and drug responses. To date, AI has driven the revolutionary advances of human organoids in life science, precision medicine and pharmaceutical science in an unprecedented way. Herein, we provide a forward-looking review, the frontiers of LOs, covering the engineered construction strategies and multidisciplinary technologies for developing LOs, highlighting the cutting-edge achievements and the prospective applications of AI in LOs, particularly in biological study, disease occurrence, disease diagnosis and prediction and drug screening in preclinical assay. Moreover, we shed light on the new research trends harnessing the power of AI for LO research in the context of multidisciplinary technologies. The aim of this paper is to motivate researchers to explore organ function throughout the human life cycle, narrow the gap between in vitro microphysiological models and the real human body, accurately predict human-related responses to external stimuli (cues and drugs), accelerate the preclinical-to-clinical transformation, and ultimately enhance the health and well-being of patients.
Collapse
Affiliation(s)
- Hui Wang
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, PR China
| | - Xiangyang Li
- Henan Engineering Research Center of Food Microbiology, College of food and bioengineering, Henan University of Science and Technology, Luoyang, 471023, PR China
- Haihe Laboratory of Synthetic Biology, Tianjin, 300308, PR China
| | - Xiaoyan You
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, PR China
- Henan Engineering Research Center of Food Microbiology, College of food and bioengineering, Henan University of Science and Technology, Luoyang, 471023, PR China
| | - Guoping Zhao
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, PR China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, PR China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, PR China
| |
Collapse
|
4
|
Lee J, Gleizes A, Janto NV, Appell LL, Sun S, Takaesu F, Webster SF, Hailstock T, Barker N, Gracz AD. Lgr5 + intestinal stem cells are required for organoid survival after genotoxic injury. Development 2024; 151:dev202941. [PMID: 39503201 PMCID: PMC11634038 DOI: 10.1242/dev.202941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024]
Abstract
Progenitors and mature cells can maintain the intestinal epithelium by dedifferentiation and facultative intestinal stem cell (fISC) function when active ISCs (aISCs) are lost to damage. Here, we modeled fISC activation in mouse intestinal organoids with doxorubicin (DXR) treatment, a chemotherapeutic known to ablate Lgr5+ aISCs in vivo. Similar fISC gene activation was observed between organoids treated with low versus high DXR, despite significantly decreased survival at the higher dose. aISCs exhibited dose-dependent loss after DXR treatment but survived at doses compatible with organoid survival. We ablated residual aISCs after DXR treatment using a Lgr52A-DTR allele and observed that aISC survival of the initial genotoxic insult is required for organoid survival following DXR treatment. These results suggest that although typical fISC genes are activated by DXR-induced injury in organoids, functional stemness remains dependent on the aISC pool. Finally, we show that human intestinal organoids require higher doses of DXR to induce loss of survival and downregulation of LGR5. Our data establish a reproducible model of DXR-induced injury in intestinal organoids and reveal differences in in vitro responses to an established in vivo damage modality.
Collapse
Affiliation(s)
- Joseph Lee
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, GA 30322, USA
| | - Antoine Gleizes
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, GA 30322, USA
| | - Nicolas V. Janto
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, GA 30322, USA
| | - Lito L. Appell
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, GA 30322, USA
| | - Siyang Sun
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, GA 30322, USA
| | - Felipe Takaesu
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA 30322, USA
| | - Sarah F. Webster
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA 30322, USA
| | - Taylor Hailstock
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA 30322, USA
| | - Nick Barker
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore 138673
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593
| | - Adam D. Gracz
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, GA 30322, USA
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, GA 30322, USA
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
5
|
Yamada M, Takezawa Y, Houry G, Düsterwald KM, Sulem D, Zhao H, Tsai YH. An Empirical Study of Self-Supervised Learning with Wasserstein Distance. ENTROPY (BASEL, SWITZERLAND) 2024; 26:939. [PMID: 39593884 PMCID: PMC11592799 DOI: 10.3390/e26110939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024]
Abstract
In this study, we consider the problem of self-supervised learning (SSL) utilizing the 1-Wasserstein distance on a tree structure (a.k.a., Tree-Wasserstein distance (TWD)), where TWD is defined as the L1 distance between two tree-embedded vectors. In SSL methods, the cosine similarity is often utilized as an objective function; however, it has not been well studied when utilizing the Wasserstein distance. Training the Wasserstein distance is numerically challenging. Thus, this study empirically investigates a strategy for optimizing the SSL with the Wasserstein distance and finds a stable training procedure. More specifically, we evaluate the combination of two types of TWD (total variation and ClusterTree) and several probability models, including the softmax function, the ArcFace probability model, and simplicial embedding. We propose a simple yet effective Jeffrey divergence-based regularization method to stabilize optimization. Through empirical experiments on STL10, CIFAR10, CIFAR100, and SVHN, we find that a simple combination of the softmax function and TWD can obtain significantly lower results than the standard SimCLR. Moreover, a simple combination of TWD and SimSiam fails to train the model. We find that the model performance depends on the combination of TWD and probability model, and that the Jeffrey divergence regularization helps in model training. Finally, we show that the appropriate combination of the TWD and probability model outperforms cosine similarity-based representation learning.
Collapse
Affiliation(s)
- Makoto Yamada
- Machine Learning and Data Science Unit, Okinawa Institute of Science and Technology, Okinawa 904-0412, Japan; (Y.T.); (G.H.); (K.M.D.)
- Center for Advanced Intelligence Project RIKEN, Tokyo 103-0027, Japan
| | - Yuki Takezawa
- Machine Learning and Data Science Unit, Okinawa Institute of Science and Technology, Okinawa 904-0412, Japan; (Y.T.); (G.H.); (K.M.D.)
- Department of Intelligence Science and Technology, Kyoto University, Kyoto 606-8501, Japan
| | - Guillaume Houry
- Machine Learning and Data Science Unit, Okinawa Institute of Science and Technology, Okinawa 904-0412, Japan; (Y.T.); (G.H.); (K.M.D.)
- Paris-Saclay Ecole Normale Superieure, 75005 Paris, France
| | - Kira Michaela Düsterwald
- Machine Learning and Data Science Unit, Okinawa Institute of Science and Technology, Okinawa 904-0412, Japan; (Y.T.); (G.H.); (K.M.D.)
- Gatsby Computational Neuroscience Unit, University College London, London WC1E 6BT, UK
| | - Deborah Sulem
- Barcelona School of Economics, Universitat Pompeu Fabra, 08002 Barcelona, Spain;
| | - Han Zhao
- Department of Computer Science, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA;
| | - Yao-Hung Tsai
- Machine Learning and Data Science Unit, Okinawa Institute of Science and Technology, Okinawa 904-0412, Japan; (Y.T.); (G.H.); (K.M.D.)
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
6
|
Russo M, Chen M, Mariella E, Peng H, Rehman SK, Sancho E, Sogari A, Toh TS, Balaban NQ, Batlle E, Bernards R, Garnett MJ, Hangauer M, Leucci E, Marine JC, O'Brien CA, Oren Y, Patton EE, Robert C, Rosenberg SM, Shen S, Bardelli A. Cancer drug-tolerant persister cells: from biological questions to clinical opportunities. Nat Rev Cancer 2024; 24:694-717. [PMID: 39223250 DOI: 10.1038/s41568-024-00737-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
The emergence of drug resistance is the most substantial challenge to the effectiveness of anticancer therapies. Orthogonal approaches have revealed that a subset of cells, known as drug-tolerant 'persister' (DTP) cells, have a prominent role in drug resistance. Although long recognized in bacterial populations which have acquired resistance to antibiotics, the presence of DTPs in various cancer types has come to light only in the past two decades, yet several aspects of their biology remain enigmatic. Here, we delve into the biological characteristics of DTPs and explore potential strategies for tracking and targeting them. Recent findings suggest that DTPs exhibit remarkable plasticity, being capable of transitioning between different cellular states, resulting in distinct DTP phenotypes within a single tumour. However, defining the biological features of DTPs has been challenging, partly due to the complex interplay between clonal dynamics and tissue-specific factors influencing their phenotype. Moreover, the interactions between DTPs and the tumour microenvironment, including their potential to evade immune surveillance, remain to be discovered. Finally, the mechanisms underlying DTP-derived drug resistance and their correlation with clinical outcomes remain poorly understood. This Roadmap aims to provide a comprehensive overview of the field of DTPs, encompassing past achievements and current endeavours in elucidating their biology. We also discuss the prospect of future advancements in technologies in helping to unveil the features of DTPs and propose novel therapeutic strategies that could lead to their eradication.
Collapse
Affiliation(s)
- Mariangela Russo
- Department of Oncology, Molecular Biotechnology Center, University of Torino, Torino, Italy.
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milano, Italy.
| | - Mengnuo Chen
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Elisa Mariella
- Department of Oncology, Molecular Biotechnology Center, University of Torino, Torino, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milano, Italy
| | - Haoning Peng
- Institute of Thoracic Oncology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Sumaiyah K Rehman
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Elena Sancho
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Alberto Sogari
- Department of Oncology, Molecular Biotechnology Center, University of Torino, Torino, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milano, Italy
| | - Tzen S Toh
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Nathalie Q Balaban
- Racah Institute of Physics, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Rene Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Matthew Hangauer
- Department of Dermatology, University of California San Diego, San Diego, CA, USA
| | | | - Jean-Christophe Marine
- Department of Oncology, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
| | - Catherine A O'Brien
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Surgery, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Yaara Oren
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - E Elizabeth Patton
- MRC Human Genetics Unit, and CRUK Scotland Centre and Edinburgh Cancer Research, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Caroline Robert
- Oncology Department, Dermatology Unit, Villejuif, France
- Oncology Department and INSERM U981, Villejuif, France
- Paris Saclay University, Villejuif, France
| | - Susan M Rosenberg
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Shensi Shen
- Institute of Thoracic Oncology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Alberto Bardelli
- Department of Oncology, Molecular Biotechnology Center, University of Torino, Torino, Italy.
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milano, Italy.
| |
Collapse
|
7
|
Qin X, Tape CJ. Functional analysis of cell plasticity using single-cell technologies. Trends Cell Biol 2024; 34:854-864. [PMID: 38355348 DOI: 10.1016/j.tcb.2024.01.006] [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: 11/02/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
Metazoan organisms are heterocellular systems composed of hundreds of different cell types, which arise from an isogenic genome through differentiation. Cellular 'plasticity' further enables cells to alter their fate in response to exogenous cues and is involved in a variety of processes, such as wound healing, infection, and cancer. Recent advances in cellular model systems, high-dimensional single-cell technologies, and lineage tracing have sparked a renaissance in plasticity research. Here, we discuss the definition of cell plasticity, evaluate state-of-the-art model systems and techniques to study cell-fate dynamics, and explore the application of single-cell technologies to obtain functional insights into cell plasticity in healthy and diseased tissues. The integration of advanced biomimetic model systems, single-cell technologies, and high-throughput perturbation studies is enabling a new era of research into non-genetic plasticity in metazoan systems.
Collapse
Affiliation(s)
- Xiao Qin
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Oxford, OX3 9DS, UK.
| | - Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London, WC1E 6DD, UK.
| |
Collapse
|
8
|
Xu X, Gao Y, Dai J, Wang Q, Wang Z, Liang W, Zhang Q, Ma W, Liu Z, Luo H, Qiao Z, Li L, Wang Z, Chen L, Zhang Y, Xiong Z. Gastric Cancer Assembloids Derived from Patient-Derived Xenografts: A Preclinical Model for Therapeutic Drug Screening. SMALL METHODS 2024; 8:e2400204. [PMID: 38948952 DOI: 10.1002/smtd.202400204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/18/2024] [Indexed: 07/02/2024]
Abstract
The construction of reliable preclinical models is crucial for understanding the molecular mechanisms involved in gastric cancer and for advancing precision medicine. Currently, existing in vitro tumor models often do not accurately replicate the human gastric cancer environment and are unsuitable for high-throughput therapeutic drug screening. In this study, droplet microfluidic technology is employed to create novel gastric cancer assembloids by encapsulating patient-derived xenograft gastric cancer cells and patient stromal cells in Gelatin methacryloyl (GelMA)-Gelatin-Matrigel microgels. The usage of GelMA-Gelatin-Matrigel composite hydrogel effectively alleviated cell aggregation and sedimentation during the assembly process, allowing for the handling of large volumes of cell-laden hydrogel and the uniform generation of assembloids in a high-throughput manner. Notably, the patient-derived xenograft assembloids exhibited high consistency with primary tumors at both transcriptomic and histological levels, and can be efficiently scaled up for preclinical drug screening efforts. Furthermore, the drug screening results clearly demonstrated that the in vitro assembloid model closely mirrored in vivo drug responses. Thus, these findings suggest that gastric cancer assembloids, which effectively replicate the in vivo tumor microenvironment, show promise for enabling more precise high-throughput drug screening and predicting the clinical outcomes of various drugs.
Collapse
Affiliation(s)
- Xinxin Xu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Medical School of Chinese PLA, Beijing, 100853, China
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yunhe Gao
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jianli Dai
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Qianqian Wang
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Zixuan Wang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Wenquan Liang
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qing Zhang
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Wenbo Ma
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Zibo Liu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Hao Luo
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Zhi Qiao
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Li Li
- Medical School of Chinese PLA, Beijing, 100853, China
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zijian Wang
- Medical School of Chinese PLA, Beijing, 100853, China
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lin Chen
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanmei Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
9
|
Cañellas-Socias A, Sancho E, Batlle E. Mechanisms of metastatic colorectal cancer. Nat Rev Gastroenterol Hepatol 2024; 21:609-625. [PMID: 38806657 DOI: 10.1038/s41575-024-00934-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2024] [Indexed: 05/30/2024]
Abstract
Despite extensive research and improvements in understanding colorectal cancer (CRC), its metastatic form continues to pose a substantial challenge, primarily owing to limited therapeutic options and a poor prognosis. This Review addresses the emerging focus on metastatic CRC (mCRC), which has historically been under-studied compared with primary CRC despite its lethality. We delve into two crucial aspects: the molecular and cellular determinants facilitating CRC metastasis and the principles guiding the evolution of metastatic disease. Initially, we examine the genetic alterations integral to CRC metastasis, connecting them to clinically marked characteristics of advanced CRC. Subsequently, we scrutinize the role of cellular heterogeneity and plasticity in metastatic spread and therapy resistance. Finally, we explore how the tumour microenvironment influences metastatic disease, emphasizing the effect of stromal gene programmes and the immune context. The ongoing research in these fields holds immense importance, as its future implications are projected to revolutionize the treatment of patients with mCRC, hopefully offering a promising outlook for their survival.
Collapse
Affiliation(s)
- Adrià Cañellas-Socias
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain.
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| | - Elena Sancho
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
10
|
Zhu H, Jin RU. The role of the fibroblast in Barrett's esophagus and esophageal adenocarcinoma. Curr Opin Gastroenterol 2024; 40:319-327. [PMID: 38626060 PMCID: PMC11155289 DOI: 10.1097/mog.0000000000001032] [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: 04/18/2024]
Abstract
PURPOSE OF REVIEW Barrett's esophagus (BE) is the number one risk factor for developing esophageal adenocarcinoma (EAC), a deadly cancer with limited treatment options that has been increasing in incidence in the US. In this report, we discuss current studies on the role of mesenchyme and cancer-associated fibroblasts (CAFs) in BE and EAC, and we highlight translational prospects of targeting these cells. RECENT FINDINGS New insights through studies using single-cell RNA sequencing (sc-RNA seq) have revealed an important emerging role of the mesenchyme in developmental signaling and cancer initiation. BE and EAC share similar stromal gene expression, as functional classifications of nonepithelial cells in BE show a remarkable similarity to EAC CAFs. Several recent sc-RNA seq studies and novel organoid fibroblast co-culture systems have characterized the subgroups of fibroblasts in BE and EAC, and have shown that these cells can directly influence the epithelium to induce BE development and cancer progression. Targeting the CAFs in EAC with may be a promising novel therapeutic strategy. SUMMARY The fibroblasts in the surrounding mesenchyme may have a direct role in influencing altered epithelial plasticity during BE development and progression to EAC.
Collapse
Affiliation(s)
- Huili Zhu
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
11
|
Fey SK, Vaquero-Siguero N, Jackstadt R. Dark force rising: Reawakening and targeting of fetal-like stem cells in colorectal cancer. Cell Rep 2024; 43:114270. [PMID: 38787726 DOI: 10.1016/j.celrep.2024.114270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/14/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Stem cells play pivotal roles in maintaining intestinal homeostasis, orchestrating regeneration, and in key steps of colorectal cancer (CRC) initiation and progression. Intriguingly, adult stem cells are reduced during many of these processes. On the contrary, primitive fetal programs, commonly detected in development, emerge during tissue repair, CRC metastasis, and therapy resistance. Recent findings indicate a dynamic continuum between adult and fetal stem cell programs. We discuss critical mechanisms facilitating the plasticity between stem cell states and highlight the heterogeneity observed upon the appearance of fetal-like states. We focus on therapeutic opportunities that arise by targeting fetal-like CRC cells and how those concepts can be translated into the clinic.
Collapse
Affiliation(s)
- Sigrid K Fey
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Nuria Vaquero-Siguero
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Rene Jackstadt
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
12
|
Zhang XY, Sui Y, Shan XF, Wang LM, Zhang L, Xie S, Cai ZG. Construction of oral squamous cell carcinoma organoids in vitro 3D-culture for drug screening. Oral Dis 2024. [PMID: 38887128 DOI: 10.1111/odi.15044] [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: 02/01/2024] [Revised: 05/06/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
OBJECTIVE Patient-derived organoids are potent pre-chemotherapy models. Due to limited research on diverse types of oral squamous cell carcinoma (OSCC) and construction efficiency, our goal was to optimize OSCC organoid models from various sites and assess drug responsiveness. METHODS We screened and optimized culture media, employing three-dimensional techniques to construct human-derived oral squamous cell carcinoma (OSCC) organoid models in vitro. Morphological validation, immunofluorescence analysis, tissue origin verification, and Short Tandem Repeat (STR) sequencing confirmed the consistency between organoids and source tissues. These organoid models were then subjected to varying concentrations of anticancer drugs, with subsequent assessment of cell viability to calculate IC50 values. RESULTS Twenty-nine surgical specimens yielded an 86.2% success rate in culturing 25 organoids in vitro. Morphological consistency confirmed nuclear atypia and positive expression of K5, P40, and E-cadherin, indicating squamous epithelial origin. Cultured complex organoids included α-SMA+ tumour-associated fibroblasts and tumour stem cells expressing CD44 and Ki67. STR sequencing affirmed genomic homogeneity between cultured organoids and source tissues. Drug sensitivity testing revealed diverse responses among organoids, highlighting their value for assessing drug sensitivity. CONCLUSIONS An efficient OSCC organoid culture system for personalized in vitro drug sensitivity screening was established, laying the foundation for precise treatment development.
Collapse
Affiliation(s)
- Xin-Yuan Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Yi Sui
- National Center for Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Department of Oral Emergency, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiao-Feng Shan
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Lu-Ming Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Lei Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Shang Xie
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Zhi-Gang Cai
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| |
Collapse
|
13
|
Xiang T, Wang J, Li H. Current applications of intestinal organoids: a review. Stem Cell Res Ther 2024; 15:155. [PMID: 38816841 PMCID: PMC11140936 DOI: 10.1186/s13287-024-03768-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
In the past decade, intestinal organoid technology has paved the way for reproducing tissue or organ morphogenesis during intestinal physiological processes in vitro and studying the pathogenesis of various intestinal diseases. Intestinal organoids are favored in drug screening due to their ability for high-throughput in vitro cultivation and their closer resemblance to patient genetic characteristics. Furthermore, as disease models, intestinal organoids find wide applications in screening diagnostic markers, identifying therapeutic targets, and exploring epigenetic mechanisms of diseases. Additionally, as a transplantable cellular system, organoids have played a significant role in the reconstruction of damaged epithelium in conditions such as ulcerative colitis and short bowel syndrome, as well as in intestinal material exchange and metabolic function restoration. The rise of interdisciplinary approaches, including organoid-on-chip technology, genome editing techniques, and microfluidics, has greatly accelerated the development of organoids. In this review, VOSviewer software is used to visualize hot co-cited journal and keywords trends of intestinal organoid firstly. Subsequently, we have summarized the current applications of intestinal organoid technology in disease modeling, drug screening, and regenerative medicine. This will deepen our understanding of intestinal organoids and further explore the physiological mechanisms of the intestine and drug development for intestinal diseases.
Collapse
Affiliation(s)
- Tao Xiang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Hui Li
- Surgical Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
14
|
Dhungel N, Dragoi AM. Exploring the multifaceted role of direct interaction between cancer cells and fibroblasts in cancer progression. Front Mol Biosci 2024; 11:1379971. [PMID: 38863965 PMCID: PMC11165130 DOI: 10.3389/fmolb.2024.1379971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/24/2024] [Indexed: 06/13/2024] Open
Abstract
The interaction between the tumor microenvironment (TME) and the cancer cells is a complex and mutually beneficial system that leads to rapid cancer cells proliferation, metastasis, and resistance to therapy. It is now recognized that cancer cells are not isolated, and tumor progression is governed among others, by many components of the TME. The reciprocal cross-talk between cancer cells and their microenvironment can be indirect through the secretion of extracellular matrix (ECM) proteins and paracrine signaling through exosomes, cytokines, and growth factors, or direct by cell-to-cell contact mediated by cell surface receptors and adhesion molecules. Among TME components, cancer-associated fibroblasts (CAFs) are of unique interest. As one of the most abundant components of the TME, CAFs play key roles in the reorganization of the extracellular matrix, facilitating metastasis and chemotherapy evasion. Both direct and indirect roles have been described for CAFs in modulating tumor progression. In this review, we focus on recent advances in understanding the role of direct contact between cancer cells and cancer-associated fibroblasts (CAFs) in driving tumor development and metastasis. We also summarize recent findings on the role of direct contact between cancer cells and CAFs in chemotherapy resistance.
Collapse
Affiliation(s)
- Nilu Dhungel
- Department of Molecular and Cellular Physiology, LSUHSC, Shreveport, LA, United States
| | - Ana-Maria Dragoi
- Department of Molecular and Cellular Physiology, LSUHSC, Shreveport, LA, United States
- Feist-Weiller Cancer Center, INLET Core, LSUHSC, Shreveport, LA, United States
| |
Collapse
|
15
|
Lee J, Gleizes A, Takaesu F, Webster SF, Hailstock T, Barker N, Gracz AD. Lgr5+ intestinal stem cells are required for organoid survival after genotoxic injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588400. [PMID: 38645040 PMCID: PMC11030406 DOI: 10.1101/2024.04.08.588400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Progenitors and mature cells can maintain the intestinal epithelium by dedifferentiation and facultative intestinal stem cell (fISC) function when active ISCs (aISCs) are lost to damage. Here, we sought to model fISC activation in intestinal organoids with doxorubicin (DXR), a chemotherapeutic known to ablate Lgr5+ aISCs in vivo. We identified low and high doses of DXR compatible with long-term organoid survival. Similar fISC gene activation was observed between organoids treated with low vs high DXR, despite significantly decreased survival at the higher dose. aISCs exhibit dose-dependent loss after DXR but survive at doses compatible with organoid survival. We ablated residual aISCs after DXR using a Lgr52A-DTR allele and observed that aISC survival of the initial genotoxic insult is required for organoid survival following DXR. These results suggest that while typical fISC genes are activated by DXR injury in organoids, functional stemness remains dependent on the aISC pool. Our data establish a reproducible model of DXR injury in intestinal organoids and reveal differences in in vitro responses to an established in vivo damage modality.
Collapse
Affiliation(s)
- Joseph Lee
- Department of Medicine, Division of Digestive Diseases, Emory University
| | - Antoine Gleizes
- Department of Medicine, Division of Digestive Diseases, Emory University
| | - Felipe Takaesu
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University
| | - Sarah F Webster
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University
| | - Taylor Hailstock
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University
| | - Nick Barker
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore
| | - Adam D Gracz
- Department of Medicine, Division of Digestive Diseases, Emory University
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University
| |
Collapse
|
16
|
Yan S, He Y, Zhu Y, Ye W, Chen Y, Zhu C, Zhan F, Ma Z. Human patient derived organoids: an emerging precision medicine model for gastrointestinal cancer research. Front Cell Dev Biol 2024; 12:1384450. [PMID: 38638528 PMCID: PMC11024315 DOI: 10.3389/fcell.2024.1384450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Gastrointestinal cancers account for approximately one-third of the total global cancer incidence and mortality with a poor prognosis. It is one of the leading causes of cancer-related deaths worldwide. Most of these diseases lack effective treatment, occurring as a result of inappropriate models to develop safe and potent therapies. As a novel preclinical model, tumor patient-derived organoids (PDOs), can be established from patients' tumor tissue and cultured in the laboratory in 3D architectures. This 3D model can not only highly simulate and preserve key biological characteristics of the source tumor tissue in vitro but also reproduce the in vivo tumor microenvironment through co-culture. Our review provided an overview of the different in vitro models in current tumor research, the derivation of cells in PDO models, and the application of PDO model technology in gastrointestinal cancers, particularly the applications in combination with CRISPR/Cas9 gene editing technology, tumor microenvironment simulation, drug screening, drug development, and personalized medicine. It also elucidates the ethical status quo of organoid research and the current challenges encountered in clinical research, and offers a forward-looking assessment of the potential paths for clinical organoid research advancement.
Collapse
Affiliation(s)
- Sicheng Yan
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuxuan He
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuehong Zhu
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wangfang Ye
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Chen
- Department of Colorectal Surgery, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Cong Zhu
- Department of Colorectal Surgery, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Fuyuan Zhan
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhihong Ma
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
17
|
Chu K, Dow LE. Adding New Dimensions to 3D Cancer Models. Cancer Res 2024; 84:798-799. [PMID: 38335538 DOI: 10.1158/0008-5472.can-24-0490] [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: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/12/2024]
Abstract
Understanding patient-specific responses to anticancer therapies and how individual tumors interact with their tumor microenvironment (TME) is a challenging task. To measure the impact of the TME on diverse and clinically relevant treatments, Ramos Zapatero and colleagues coupled patient-derived organoid (PDO) and cancer-associated fibroblast (CAF) cocultures with high-throughput mass cytometry-based assessment of cell state. Using a newly developed "Trellis" algorithm enabled integration and analysis of highly complex, multidimensional treatment response data. This work showed that tumor cell response to chemotherapy was associated with both intrinsic and nonintrinsic signaling states, whereby proliferative rate, growth factor signaling, and CAFs interaction influenced chemoprotection. Furthermore, the work suggests a potential role for the TME in promoting lineage plasticity associated with drug resistance. In all, the pipeline described provides a blueprint for exploring the intricate interplay of factors influencing cancer treatment response.
Collapse
Affiliation(s)
- Kevan Chu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, New York
| | - Lukas E Dow
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, New York
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Department of Biochemistry, Weill Cornell Medicine, New York, New York
| |
Collapse
|
18
|
Ogasawara N, Kano Y, Yoneyama Y, Kobayashi S, Watanabe S, Kirino S, Velez-Bravo FD, Hong Y, Ostapiuk A, Lutsik P, Onishi I, Yamauchi S, Hiraguri Y, Ito G, Kinugasa Y, Ohashi K, Watanabe M, Okamoto R, Tejpar S, Yui S. Discovery of non-genomic drivers of YAP signaling modulating the cell plasticity in CRC tumor lines. iScience 2024; 27:109247. [PMID: 38439969 PMCID: PMC10910304 DOI: 10.1016/j.isci.2024.109247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 03/06/2024] Open
Abstract
In normal intestines, a fetal/regenerative/revival cell state can be induced upon inflammation. This plasticity in cell fate is also one of the current topics in human colorectal cancer (CRC). To dissect the underlying mechanisms, we generated human CRC organoids with naturally selected genetic mutation profiles and exposed them to two different conditions by modulating the extracellular matrix (ECM). Among tested mutation profiles, a fetal/regenerative/revival state was induced following YAP activation via a collagen type I-enriched microenvironment. Mechanistically, YAP transcription was promoted by activating AP-1 and TEAD-dependent transcription and suppressing intestinal lineage-determining transcription via mechanotransduction. The phenotypic conversion was also involved in chemoresistance, which could be potentially resolved by targeting the underlying YAP regulatory elements, a potential target of CRC treatment.
Collapse
Affiliation(s)
- Nobuhiko Ogasawara
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yoshihito Kano
- Department of Clinical Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yosuke Yoneyama
- Institute of Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Sakurako Kobayashi
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Satoshi Watanabe
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Sakura Kirino
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | | | - Yourae Hong
- Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Pavlo Lutsik
- Computational Cancer Biology and Epigenomics, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Iichiroh Onishi
- Department of Diagnostic Pathology, Tokyo Medical and Dental University Hospital, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Shinichi Yamauchi
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yui Hiraguri
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Go Ito
- Advanced Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yusuke Kinugasa
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Kenichi Ohashi
- Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Mamoru Watanabe
- Advanced Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Sabine Tejpar
- Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Shiro Yui
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| |
Collapse
|
19
|
Tape CJ. Plastic persisters: revival stem cells in colorectal cancer. Trends Cancer 2024; 10:185-195. [PMID: 38071119 DOI: 10.1016/j.trecan.2023.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 03/16/2024]
Abstract
Colorectal cancer (CRC) is traditionally considered to be a genetically driven disease. However, nongenetic plasticity has recently emerged as a major driver of tumour initiation, metastasis, and therapy response in CRC. Central to these processes is a recently discovered cell type, the revival colonic stem cell (revCSC). In contrast to traditional proliferative CSCs (proCSCs), revCSCs prioritise survival over propagation. revCSCs play an essential role in primary tumour formation, metastatic dissemination, and nongenetic chemoresistance. Current evidence suggests that CRC tumours leverage intestinal stem cell plasticity to both proliferate (via proCSCs) when unchallenged and survive (via revCSCs) in response to cell-extrinsic pressures. Although revCSCs likely represent a major source of therapeutic failure in CRC, our increasing knowledge of this important stem cell fate provides novel opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London, WC1E 6DD, UK.
| |
Collapse
|
20
|
Qin X, Cardoso Rodriguez F, Sufi J, Vlckova P, Claus J, Tape CJ. An oncogenic phenoscape of colonic stem cell polarization. Cell 2023; 186:5554-5568.e18. [PMID: 38065080 DOI: 10.1016/j.cell.2023.11.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/14/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023]
Abstract
Cancer cells are regulated by oncogenic mutations and microenvironmental signals, yet these processes are often studied separately. To functionally map how cell-intrinsic and cell-extrinsic cues co-regulate cell fate, we performed a systematic single-cell analysis of 1,107 colonic organoid cultures regulated by (1) colorectal cancer (CRC) oncogenic mutations, (2) microenvironmental fibroblasts and macrophages, (3) stromal ligands, and (4) signaling inhibitors. Multiplexed single-cell analysis revealed a stepwise epithelial differentiation phenoscape dictated by combinations of oncogenes and stromal ligands, spanning from fibroblast-induced Clusterin (CLU)+ revival colonic stem cells (revCSCs) to oncogene-driven LRIG1+ hyper-proliferative CSCs (proCSCs). The transition from revCSCs to proCSCs is regulated by decreasing WNT3A and TGF-β-driven YAP signaling and increasing KRASG12D or stromal EGF/Epiregulin-activated MAPK/PI3K flux. We find that APC loss and KRASG12D collaboratively limit access to revCSCs and disrupt stromal-epithelial communication-trapping epithelia in the proCSC fate. These results reveal that oncogenic mutations dominate homeostatic differentiation by obstructing cell-extrinsic regulation of cell-fate plasticity.
Collapse
Affiliation(s)
- Xiao Qin
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Ferran Cardoso Rodriguez
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Jahangir Sufi
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Petra Vlckova
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Jeroen Claus
- Phospho Biomedical Animation, The Greenhouse Studio 6, London N17 9QU, UK
| | - Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK.
| |
Collapse
|