1
|
Abed H, Radha R, Anjum S, Paul V, AlSawaftah N, Pitt WG, Ashammakhi N, Husseini GA. Targeted Cancer Therapy-on-A-Chip. Adv Healthc Mater 2024; 13:e2400833. [PMID: 39101627 PMCID: PMC11582519 DOI: 10.1002/adhm.202400833] [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: 03/04/2024] [Revised: 06/15/2024] [Indexed: 08/06/2024]
Abstract
Targeted cancer therapy (TCT) is gaining increased interest because it reduces the risks of adverse side effects by specifically treating tumor cells. TCT testing has traditionally been performed using two-dimensional (2D) cell culture and animal studies. Organ-on-a-chip (OoC) platforms have been developed to recapitulate cancer in vitro, as cancer-on-a-chip (CoC), and used for chemotherapeutics development and testing. This review explores the use of CoCs to both develop and test TCTs, with a focus on three main aspects, the use of CoCs to identify target biomarkers for TCT development, the use of CoCs to test free, un-encapsulated TCTs, and the use of CoCs to test encapsulated TCTs. Despite current challenges such as system scaling, and testing externally triggered TCTs, TCToC shows a promising future to serve as a supportive, pre-clinical platform to expedite TCT development and bench-to-bedside translation.
Collapse
Affiliation(s)
- Heba Abed
- Department of Chemical and Biological EngineeringAmerican University of SharjahSharjahUAE
| | - Remya Radha
- Department of Chemical and Biological EngineeringAmerican University of SharjahSharjahUAE
| | - Shabana Anjum
- Department of Chemical and Biological EngineeringAmerican University of SharjahSharjahUAE
| | - Vinod Paul
- Materials Science and Engineering PhD programCollege of Arts and SciencesAmerican University of SharjahSharjahUAE
| | - Nour AlSawaftah
- Materials Science and Engineering PhD programCollege of Arts and SciencesAmerican University of SharjahSharjahUAE
| | - William G. Pitt
- Department of Chemical EngineeringBrigham Young UniversityProvoUT84602USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME)Michigan State UniversityEast LansingMI48824USA
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095‐1600USA
| | - Ghaleb A. Husseini
- Department of Chemical and Biological EngineeringAmerican University of SharjahSharjahUAE
- Materials Science and Engineering PhD programCollege of Arts and SciencesAmerican University of SharjahSharjahUAE
| |
Collapse
|
2
|
Chen LY, Chou YT, Liew PL, Chu LH, Wen KC, Lin SF, Weng YC, Wang HC, Su PH, Lai HC. In vitro drug testing using patient-derived ovarian cancer organoids. J Ovarian Res 2024; 17:194. [PMID: 39358778 PMCID: PMC11445862 DOI: 10.1186/s13048-024-01520-2] [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: 04/22/2024] [Accepted: 09/21/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Ovarian cancer is the most lethal gynecological cancer. As the primary treatment, chemotherapy has a response rate of only 60-70% in advanced stages, and even lower as a second-line treatment. Despite guideline recommendations, which drugs will be most effective remains unclear. Thus, a strategy to prioritize chemotherapy options is urgently needed. Cancer organoids have recently emerged as a method for in vitro drug testing. However, limited clinical correlations have been assessed with test results from cancer organoids, particularly in gynecological cancers. We therefore aimed to generate patient-derived organoids (PDOs) of ovarian cancer, to assess their drug sensitivities and correlations with patient clinical outcomes. METHODS PDOs were generated from fresh tumors obtained during surgical resection, which was then cultured under matrix gel and appropriate growth factors. Morphological and molecular characterization of PDOs were assessed by phase contrast microscopy and paraffin-embedded histopathology. Expressions of PAX8, TP53, WT1, CK7, and CK20 were tested by immunohistochemical staining and compared with parental tumor tissues and the human protein atlas database. PDOs were subjected to in vitro drug testing to determine drug sensitivity using Titer-Glo® 3D Cell Viability Assay. PDO viability was measured, and area under the curve calculated, to compare responses to various compounds. Correlations were calculated between selected patients' clinical outcomes and in vitro drug testing results. RESULTS We established 31 PDOs. Among them, 28 PDOs can be expanded, including 15, 11, and 2 from ovarian, endometrial, and cervical cancers, respectively. The PDOs preserved the histopathological profiles of their originating tumors. In vitro drug testing of 10 ovarian cancer PDOs revealed individual differential responses to recommended drugs, and interpersonal heterogeneity in drug sensitivity, even with the same histology type. Among four patients who were platinum sensitive, resistant, or refractory, PDO drug responses correlated well with their clinical courses. CONCLUSION In vitro drug testing using ovarian cancer organoids is feasible and correlates well with patient clinical responses. These results may facilitate development of precision chemotherapy and personalized screening for repurposed or new drugs.
Collapse
Affiliation(s)
- Lin-Yu Chen
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Yu-Ting Chou
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Phui-Ly Liew
- Department of Pathology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Ling-Hui Chu
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Kuo-Chang Wen
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Shiou-Fu Lin
- Department of Pathology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Yu-Chun Weng
- Translational Epigenetics Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Hui-Chen Wang
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Po-Hsuan Su
- College of Health Technology, National Taipei University of Nursing and Health Sciences, Taipei, 11219, Taiwan
| | - Hung-Cheng Lai
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan.
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Translational Epigenetics Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan.
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan.
| |
Collapse
|
3
|
Li J, Zhou M, Xie J, Chen J, Yang M, Ye C, Cheng S, Liu M, Li R, Tan R. Organoid modeling meets cancers of female reproductive tract. Cell Death Discov 2024; 10:410. [PMID: 39333482 PMCID: PMC11437045 DOI: 10.1038/s41420-024-02186-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
Diseases of the female reproductive system, especially malignant tumors, pose a serious threat to women's health worldwide. One of the key factors limiting research progress in this area is the lack of representative models. Organoid technology, especially tumor organoids, has been increasingly applied in the study of female reproductive system tumors due to their high heterogeneity, close resemblance to the physiological state, easy acquisition and cultivation advantages. They play a significant role in understanding the origin and causes of tumors, drug screening, and personalized treatment and more. This article reviews the organoid models for the female reproductive system, focusing on the cancer research advancements. It discusses the methods for constructing tumor organoids of the female reproductive tract and summarizes the limitations of current research. The aim is to offer a reference for future development and application of these organoid models, contributing to the advancement of anti-tumor drugs and treatment strategies for female reproductive tract cancer patients.
Collapse
Affiliation(s)
- Jiao Li
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Mengting Zhou
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Xie
- Information Technology Center, West China Hospital of Sichuan University, Sichuan University, Chengdu, China
| | - Jiani Chen
- Chongqing Medical University, Chongqing, China
| | - Mengni Yang
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Changjun Ye
- Rehabilitation Department, Changgeng Yining Hospital, Wenzhou, China
| | - Shihu Cheng
- Geriatric Department, Changgeng Yining Hospital, Wenzhou, China
| | - Miao Liu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| |
Collapse
|
4
|
Zhao Y, Wang C, Deng W, Li L, Liu J, Shi Y, Tao X, Zhang J, Cao Q, Cai C, Han X. Patient-derived ovarian cancer organoid carries immune microenvironment and blood vessel keeping high response to cisplatin. MedComm (Beijing) 2024; 5:e697. [PMID: 39206413 PMCID: PMC11351687 DOI: 10.1002/mco2.697] [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: 11/08/2023] [Revised: 06/15/2024] [Accepted: 06/28/2024] [Indexed: 09/04/2024] Open
Abstract
Ovarian cancer is high recurrence and mortality malignant tumor. The most common ovarian cancer was High-Grade Serous Ovarian Cancer. However, High-Grade Serous Ovarian Cancer organoid is rare, which organoid with patient immune microenvironment and blood vessels even absence. Here, we report a novel High-Grade Serous Ovarian Cancer organoid system derived from patient ovarian cancer samples. These organoids recapitulate High-Grade Serous Ovarian Cancer organoids' histological and molecular heterogeneity while preserving the critical immune microenvironment and blood vessels, as evidenced by the presence of CD34 + endothelial cells. Whole exome sequencing identifies key mutations (CSMD3, TP53, GABRA6). Organoids show promise in testing cisplatin sensitivity for patients resistant to carboplatin and paclitaxel, with notable responses in cancer proteoglycans and p53 (TP53) signaling, like ACTG/ACTB1/AKT2 genes and BBC3/MDM2/PERP. Integration of immune microenvironment and blood vessels enhances potential for novel therapies like immunotherapies and angiogenesis inhibitors. Our work may provide a new detection system and theoretical basis for ovarian cancer research and individual therapy.
Collapse
Affiliation(s)
- Yuqing Zhao
- Obstetrics & Gynecology HospitalFudan UniversityShanghaiChina
| | - Chen Wang
- Department of ResearchShanghai LiSheng BiotechShanghaiChina
| | - Wei Deng
- LongHua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
- Department of OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lanyang Li
- Department of ResearchShanghai LiSheng BiotechShanghaiChina
| | - Jiping Liu
- Department of ResearchShanghai LiSheng BiotechShanghaiChina
| | - Yanghua Shi
- Department of ResearchShanghai LiSheng BiotechShanghaiChina
| | - Xiang Tao
- Obstetrics & Gynecology HospitalFudan UniversityShanghaiChina
| | - Jian Zhang
- Department of ResearchShanghai LiSheng BiotechShanghaiChina
| | - Qi Cao
- Obstetrics & Gynecology HospitalFudan UniversityShanghaiChina
| | - Chunhui Cai
- Department of ResearchShanghai LiSheng BiotechShanghaiChina
| | - Xinxin Han
- Department of ResearchShanghai LiSheng BiotechShanghaiChina
- Organ Regeneration X LabLiSheng East China Institute of BiotechnologyPeking UniversityJiangsuChina
| |
Collapse
|
5
|
Cortesi M, Warton K, Ford CE. Beyond 2D cell cultures: how 3D models are changing the in vitro study of ovarian cancer and how to make the most of them. PeerJ 2024; 12:e17603. [PMID: 39221267 PMCID: PMC11366228 DOI: 10.7717/peerj.17603] [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: 04/10/2024] [Accepted: 05/30/2024] [Indexed: 09/04/2024] Open
Abstract
3D cell cultures are a fundamental tool in ovarian cancer research that can enable more effective study of the main features of this lethal disease, including the high rates of recurrence and chemoresistance. A clearer, more comprehensive understanding of the biological underpinnings of these phenomena could aid the development of more effective treatments thus improving patient outcomes. Selecting the most appropriate model to investigate the different aspects of cell biology that are relevant to cancer is challenging, especially since the assays available for the study of 3D cultures are not fully established yet. To maximise the usefulness of 3D cell cultures of ovarian cancer, we undertook an in-depth review of the currently available models, taking into consideration the strengths and limitations of each approach and of the assay techniques used to evaluate the results. This integrated analysis provides insight into which model-assay pair is best suited to study different parameters of ovarian cancer biology such as cell proliferation, gene expression or treatment response. We also describe how the combined use of multiple models is likely to be the most effective strategy for the in vitro characterisation of complex behaviours.
Collapse
Affiliation(s)
- Marilisa Cortesi
- School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Department of Electrical Electronic and Information Engineering “G. Marconi”, University of Bologna, Cesena, Italy
| | - Kristina Warton
- School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Caroline E. Ford
- School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
6
|
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
|
7
|
Ding H, Hu B, Guo R. Comprehensive analysis of single cell and bulk data develops a promising prognostic signature for improving immunotherapy responses in ovarian cancer. PLoS One 2024; 19:e0298125. [PMID: 38346070 PMCID: PMC10861092 DOI: 10.1371/journal.pone.0298125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/19/2024] [Indexed: 02/15/2024] Open
Abstract
The tumor heterogeneity is an important cause of clinical therapy failure and yields distinct prognosis in ovarian cancer (OV). Using the advantages of integrated single cell RNA sequencing (scRNA-seq) and bulk data to decode tumor heterogeneity remains largely unexplored. Four public datasets were enrolled in this study, including E-MTAB-8107, TCGA-OV, GSE63885, and GSE26193 cohorts. Random forest algorithm was employed to construct a multi-gene prognostic panel and further evaluated by receiver operator characteristic (ROC), calibration curve, and Cox regression. Subsequently, molecular characteristics were deciphered, and treatments strategies were explored to deliver precise therapy. The landscape of cell subpopulations and functional characteristics, as well as the dynamic of macrophage cells were detailly depicted at single cell level, and then screened prognostic candidate genes. Based on the expression of candidate genes, a stable and robust cell characterized gene associated prognosis signature (CCIS) was developed, which harbored excellent performance at prognosis assessment and patient stratification. The ROC and calibration curves, and Cox regression analysis elucidated CCIS could serve as serve as an independent factor for predicting prognosis. Moreover, a promising clinical tool nomogram was also constructed according to stage and CCIS. Through comprehensive investigations, patients in low-risk group were charactered by favorable prognosis, elevated genomic variations, higher immune cell infiltrations, and superior antigen presentation. For individualized treatment, patients in low-risk group were inclined to better immunotherapy responses. This study dissected tumor heterogeneity and afforded a promising prognostic signature, which was conducive to facilitating clinical outcomes for patients with OV.
Collapse
Affiliation(s)
- Huanfei Ding
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, China
| | - Bowen Hu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruixia Guo
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, China
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Choi MS, Lee CY, Kim JH, Lee YM, Lee S, Kim HJ, Heo K. Gramicidin, a Bactericidal Antibiotic, Is an Antiproliferative Agent for Ovarian Cancer Cells. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:2059. [PMID: 38138162 PMCID: PMC10744341 DOI: 10.3390/medicina59122059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/25/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023]
Abstract
Background and Objectives: Gramicidin, a bactericidal antibiotic used in dermatology and ophthalmology, has recently garnered attention for its inhibitory actions against cancer cell growth. However, the effects of gramicidin on ovarian cancer cells and the underlying mechanisms are still poorly understood. We aimed to elucidate the anticancer efficacy of gramicidin against ovarian cancer cells. Materials and Methods: The anticancer effect of gramicidin was investigated through an in vitro experiment. We analyzed cell proliferation, DNA fragmentation, cell cycle arrest and apoptosis in ovarian cancer cells using WST-1 assay, terminal deoxynucleotidyl transferase dUTP nick and labeling (TUNEL), DNA agarose gel electrophoresis, flow cytometry and western blot. Results: Gramicidin treatment induces dose- and time-dependent decreases in OVCAR8, SKOV3, and A2780 ovarian cancer cell proliferation. TUNEL assay and DNA agarose gel electrophoresis showed that gramicidin caused DNA fragmentation in ovarian cancer cells. Flow cytometry demonstrated that gramicidin induced cell cycle arrest. Furthermore, we confirmed via Western blot that gramicidin triggered apoptosis in ovarian cancer cells. Conclusions: Our results strongly suggest that gramicidin exerts its inhibitory effect on cancer cell growth by triggering apoptosis. Conclusively, this study provides new insights into the previously unexplored anticancer properties of gramicidin against ovarian cancer cells.
Collapse
Affiliation(s)
- Min Sung Choi
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (M.S.C.); (Y.M.L.); (S.L.)
| | - Chae Yeon Lee
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (C.Y.L.); (J.H.K.)
| | - Ji Hyeon Kim
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (C.Y.L.); (J.H.K.)
| | - Yul Min Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (M.S.C.); (Y.M.L.); (S.L.)
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (M.S.C.); (Y.M.L.); (S.L.)
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (C.Y.L.); (J.H.K.)
- Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| | - Hyun Jung Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (M.S.C.); (Y.M.L.); (S.L.)
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (C.Y.L.); (J.H.K.)
- Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| | - Kyun Heo
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (M.S.C.); (Y.M.L.); (S.L.)
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (C.Y.L.); (J.H.K.)
- Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
9
|
Guler Kara H, Ozates NP, Asik A, Gunduz C. Cancer stemness kinase inhibitor amcasertib: a promising therapeutic agent in ovarian cancer stem and cancer cell models with different genetic profiles. Med Oncol 2023; 40:342. [PMID: 37891391 DOI: 10.1007/s12032-023-02210-7] [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: 09/04/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023]
Abstract
Ovarian cancer, often referred to as the 'silent killer,' is a significant contributor to mortality rates. Emerging evidence implicates Nanog as a potential therapeutic target in ovarian cancer. Amcasertib (BBI-503) is an orally administered primary class stemness kinase inhibitor that effectively targets NANOG and various cancer stem cell pathways by specifically inhibiting serine-threonine stemness kinases. This study aimed to evaluate the antineoplastic effects of Nanog inhibition, a critical transcription factor associated with pluripotency and its role in ovarian cancer tumorigenesis, using the novel therapeutic agent Amcasertib in ovarian cancer cells characterized by distinct genetic profiles. The cytotoxicity of Amcasertib was assessed in both ovarian cancer and cancer stem cell models utilizing the Xelligence-RTCA system. The impact of the determined IC50 dose on apoptosis, invasion, migration, epithelial-mesenchymal transition (EMT), cell cycle progression, colony formation, and spheroid growth was evaluated using appropriate analytical techniques. Our findings revealed that Amcasertib exhibited significant antiproliferative effects and induced apoptosis in ovarian cancer and cancer stem cells. Moreover, Amcasertib caused G1 phase arrest and impeded colony formation in MDAH-2774 cells. Additionally, Amcasertib effectively inhibited spheroid growth in OVCAR-3 and OCSC cells. Notably, it demonstrated the ability to suppress invasion and migration in MDAH-2774 and OCSC cells. Furthermore, the suppression of Nanog-mediated stem cell-like features by Amcasertib was particularly pronounced in ER-negative ovarian cancer and cancer stem cells, highlighting its high anticancer efficacy in this subgroup. These results suggest that Amcasertib holds promise as a potential standalone or combination therapy agent for the treatment of ER-negative ovarian cancer.
Collapse
Affiliation(s)
- Hale Guler Kara
- Department of Medical Biology, Medical Faculty, Harran University, Haliliye, 63050, Gülveren, Sanliurfa, Turkey.
| | - Neslihan Pinar Ozates
- Department of Medical Biology, Medical Faculty, Harran University, Haliliye, 63050, Gülveren, Sanliurfa, Turkey
| | - Aycan Asik
- Department of Medical Biology, Medical Faculty, Mugla Sitki Kocman University, Menteşe, 48000, Muğla, Turkey
| | - Cumhur Gunduz
- Department of Medical Biology, Medical Faculty, Ege University, Bornova, 35100, Izmir, Turkey
| |
Collapse
|
10
|
Chan WS, Mo X, Ip PPC, Tse KY. Patient-derived organoid culture in epithelial ovarian cancers-Techniques, applications, and future perspectives. Cancer Med 2023; 12:19714-19731. [PMID: 37776168 PMCID: PMC10587945 DOI: 10.1002/cam4.6521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/05/2023] [Accepted: 08/30/2023] [Indexed: 10/01/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is a heterogeneous disease composed of different cell types with different molecular aberrations. Traditional cell lines and mice models cannot recapitulate the human tumor biology and tumor microenvironment (TME). Patient-derived organoids (PDOs) are freshly derived from patients' tissues and are then cultured with extracellular matrix and conditioned medium. The high concordance of epigenetic, genomic, and proteomic landscapes between the parental tumors and PDOs suggests that PDOs can provide more reliable results in studying cancer biology, allowing high throughput drug screening, and identifying their associated signaling pathways and resistance mechanisms. However, despite having a heterogeneity of cells in PDOs, some cells in TME will be lost during the culture process. Next-generation organoids have been developed to circumvent some of the limitations. Genetically engineered organoids involving targeted gene editing can facilitate the understanding of tumorigenesis and drug response. Co-culture systems where PDOs are cultured with different cell components like immune cells can allow research using immunotherapy which is otherwise impossible in conventional cell lines. In this review, the limitations of the traditional in vitro and in vivo assays, the use of PDOs, the challenges including some tips and tricks of PDO generation in EOC, and the future perspectives, will be discussed.
Collapse
Affiliation(s)
- Wai Sun Chan
- Department of Obstetrics and GynaecologyThe University of Hong KongPokfulamHong Kong SAR
| | - Xuetang Mo
- Department of Obstetrics and GynaecologyThe University of Hong KongPokfulamHong Kong SAR
| | | | - Ka Yu Tse
- Department of Obstetrics and GynaecologyThe University of Hong KongPokfulamHong Kong SAR
| |
Collapse
|
11
|
Kumar S, Raina M, Tankay K, Ingle GM. Patient-derived organoids in ovarian cancer: Current research and its clinical relevance. Biochem Pharmacol 2023; 213:115589. [PMID: 37196684 DOI: 10.1016/j.bcp.2023.115589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/19/2023]
Abstract
Regardless of recent advances in cancer treatment, ovarian cancer (OC) patients have had a five-year survival rate of 48% in the last few decades. Diagnosis at the advanced stage, disease recurrence, and lack of early biomarkers are the severe clinical challenges associated with disease survival rate. Identifying tumor origin and developing precision drugs will effectively advance OC patient's treatment. The lack of a proper platform to identify and develop new therapeutic strategies in OC treatment necessitates searching for a suitable model to address tumor recurrence and therapeutic resistance. The development of the OC patient-derived organoid model provided a unique platform to identify the exact origin of high-grade serous OC, drug screening, and the development of precision medicine. This review provides an overview of recent progress in developing patient-derived organoids and their clinical relevance. Here, we outline their uses for transcriptomics and genomics profiling, drug screening, translational study, and their future perspective and clinical outlook as a model to advance OC research that could offer a promising approach for developing precision medicine.
Collapse
Affiliation(s)
- Sanjay Kumar
- Division of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, India.
| | - Manita Raina
- Division of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, India
| | - Kalpana Tankay
- Division of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, India
| | - Gaurav Milind Ingle
- Division of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, India
| |
Collapse
|
12
|
Yang L, Gu Y. SPTBN2 regulates endometroid ovarian cancer cell proliferation, invasion and migration via ITGB4‑mediated focal adhesion and ECM receptor signalling pathway. Exp Ther Med 2023; 25:277. [PMID: 37206547 PMCID: PMC10189743 DOI: 10.3892/etm.2023.11977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/31/2023] [Indexed: 05/21/2023] Open
Abstract
Ovarian cancer is as a major contributor to gynaecologic death globally. The present study aimed to investigate the regulatory role of spectrin β non-erythrocytic 2 gene (SPTBN2) in endometroid ovarian cancer and its mechanism of action. According to the Gene Expression Profiling Interactive Analysis (GEPIA) database, SPTBN2 expression is elevated in ovarian cancer tissues and higher SPTBN2 expression indicated a worse prognosis. The present study assessed SPTBN2 mRNA and protein expression levels by reverse transcription-quantitative PCR and western blotting, respectively. Cell viability, proliferation, migration and invasion were assessed with Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine incorporation, wound healing and Transwell assays, respectively. SPTBN2 expression was notably enhanced in ovarian cancer cell lines, especially in A2780 cells compared with HOSEPiC cells (P<0.001). Following transfection with small interfering (si)RNA targeting SPTBN2, the viability, proliferation, migration and invasion of A2780 cells were decreased compared with those of A2780 cells transfected with siRNA-NC (P<0.001). Gene Set Enrichment Analysis database revealed that SPTBN2 was primarily enriched in 'focal adhesion' and 'extracellular matrix (ECM)-receptor interaction', whereas SPTBN2 was significantly associated with integrin β4 (ITGB4) in the GEPIA database. In addition, rescue experiments were performed to determine the mechanism of SPTBN2 in endometroid ovarian cancer. ITGB4 overexpression reversed the inhibitory effects of the SPTBN2 knockdown on viability, proliferation, migration and invasion of A2780 cells (P<0.05). The impacts of SPTBN2 on the expression of focal adhesion and downstream ECM receptor signalling-related proteins, including Src and p-FAK/FAK, were significantly reversed by ITGB4 overexpression (P<0.01). Collectively, SPTBN2 may regulate endometroid ovarian cancer cell proliferation, invasion and migration through the ITGB4-mediated focal adhesion and ECM receptor signalling pathway.
Collapse
Affiliation(s)
- La Yang
- Department of Obstetrics and Gynaecology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, P.R. China
- Correspondence to: Dr La Yang, Department of Obstetrics and Gynaecology, Affiliated Hospital of Guizhou Medical University, 28 Guiyi Street, Guiyang, Guizhou 550001, P.R. China
| | - Yuanyuan Gu
- Department of Obstetrics and Gynaecology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, P.R. China
| |
Collapse
|
13
|
Hu P, Lei L, Wang Y, Tian X, Wei X, Jiang N, Liu L. CLDN4 as a Novel Diagnostic and Prognostic Biomarker and Its Association with Immune Infiltrates in Ovarian Cancer. Mediators Inflamm 2023; 2023:1075265. [PMID: 37057131 PMCID: PMC10089777 DOI: 10.1155/2023/1075265] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/05/2023] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
Ovarian cancer (OC) is the seventh most prevalent type of cancer in women and the second most common cause of cancer-related deaths in women worldwide. Because of the high rates of relapse, there is an immediate and pressing need for the discovery of innovative sensitive biomarkers for OC patients. Using TCGA and GSE26712 datasets, we were able to identify 17 survival-related DEGs in OC that had differential expression. CLDN4 was the gene that caught our attention the most out of the 17 important genes since its expression was much higher in OC samples than in nontumor samples. The findings of the ROC assays then confirmed the diagnostic utility of the test in screening OC specimens to differentiate them from nontumor specimens. Patients with high CLDN4 expression predicted a shorter overall survival (OS) and disease-specific survival (DSS) than those with low CLDN4 expression, according to clinical research. Patients with low CLDN4 expression predicted longer OS and DSS. Analysis using both univariate and multivariate techniques revealed that CLDN4 expression was an independent factor associated with a poor prognosis for OS and DSS. Based on multivariate analysis, the
-indexes and calibration plots of the nomogram suggested an effective predictive performance for OC patients. After that, we investigated whether or not there was a link between the infiltration of immune cells and the expression of the CLDN4 gene. We found that the expression of CLDN4 was positively associated with Th17 cells, NK CD56bright cells, while negatively associated with Th2 cells, pDC, and T helper cells. In the end, we carried out RT-PCR on our cohort and confirmed that the level of CLDN4 expression was noticeably elevated in OC specimens in comparison to nontumor tissues. The diagnostic usefulness of CLDN4 expression for OC was also validated by the findings of ROC tests. Thus, our findings revealed that CLDN4 may serve as a predictive biomarker in OC to assess both the clinical outcome of OC patients and the level of immune infiltration.
Collapse
Affiliation(s)
- Pan Hu
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, China
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Li Lei
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, China
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, China
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xue Tian
- Department of Gynecology, Women and Children’s Hospital of Xiushan County, Chongqing, China
| | - Xia Wei
- People’s Hospital of Kunming, Xishan District, Kunming, Yunnan, China
| | - Ni Jiang
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, China
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Lubin Liu
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, China
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
14
|
Li G, Xu W, Li X, Chen M, Shi Y, Wei M, Peng D. Oncogenic SIRT7 inhibits GATA4 transcriptional activity and activates the Wnt signaling pathway in ovarian cancer. Gynecol Oncol 2023; 171:39-48. [PMID: 36804620 DOI: 10.1016/j.ygyno.2022.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 02/19/2023]
Abstract
OBJECTIVE Sirtuin-7 (SIRT7) is a class III histone deacetylase that plays an important role in cancer development and frequently overexpressed in carcinomas. In this study, the tumor-supporting role and underlying mechanisms of SIRT7 were characterized in ovarian cancer (OC) aggressiveness. METHODS SIRT7 expression was examined in OC tissues and cells. Interactions among SIRT7, GATA4, Wnt signaling pathway were explored by bioinformatics tools and experimental validations. The effect of SIRT7 and GATA4 on malignant phenotypes of OC cells were examined with gain- and loss-of-function experiments. A nude mouse model of OC was developed to verify the in vitro findings. RESULTS It was noted that SIRT7 was highly expressed in OC tissues and cells. Cell lines with higher SIRT7 expression (OVCAR-3 and OVCAR-8) were used for subsequent in vitro experiments. The experimental data indicated that silencing of SIRT7 suppressed the OC cell proliferation, colony formation, migration, and invasion, and promoted cell senescence, which could be abolished by GATA4 knockdown. Mechanistically, SIRT7 promoted deacetylation of GATA4 and consequently inhibited the transcriptional activity of GATA4. In addition, GATA4 induced OC cell senescence by inhibiting Wnt signaling pathway. Further in vivo experiments substantiated that SIRT7 knockdown or overexpressed GATA4 could effectively inhibit tumor growth of nude mice. CONCLUSION Taken together, our findings indicated that SIRT7 enhanced development of OC by suppressing GATA4 and activating Wnt signaling pathway, suggesting the potential of SIRT7/GATA4/Wnt axis as a therapeutic target for OC.
Collapse
Affiliation(s)
- Guocheng Li
- Southeast University School of Medicine, Nanjing 210009, PR China
| | - Wenwen Xu
- Southeast University School of Medicine, Nanjing 210009, PR China
| | - Xuelin Li
- Department of Obstetrics and Gynecology, Zhongda Hospital Southeast University, Nanjing 210009, PR China
| | - Mengzhu Chen
- Department of Obstetrics and Gynecology, Zhongda Hospital Southeast University, Nanjing 210009, PR China
| | - Yong Shi
- Department of Obstetrics and Gynecology, Zhongda Hospital Southeast University, Nanjing 210009, PR China
| | - Mingming Wei
- Southeast University School of Medicine, Nanjing 210009, PR China
| | - Danhong Peng
- Department of Obstetrics and Gynecology, Zhongda Hospital Southeast University, Nanjing 210009, PR China.
| |
Collapse
|
15
|
Jun HR, Kang HJ, Ju SH, Kim JE, Jeon SY, Ku B, Lee JJ, Kim M, Kim MJ, Choi JJ, Noh JJ, Kim HS, Lee JW, Lee JK, Lee DW. High-throughput organo-on-pillar (high-TOP) array system for three-dimensional ex vivo drug testing. Biomaterials 2023; 296:122087. [PMID: 36924663 DOI: 10.1016/j.biomaterials.2023.122087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/05/2023] [Indexed: 03/09/2023]
Abstract
The development of organoid culture technologies has triggered industrial interest in ex vivo drug test-guided clinical response prediction for precision cancer therapy. The three-dimensional culture encapsulated with basement membrane (BM) components is extremely important in establishing ex vivo organoids and drug sensitivity tests because the BM components confer essential structures resembling tumor histopathology. Although numerous studies have demonstrated three-dimensional culture-based drug screening methods, establishing a large-scale drug-screening platform with matrix-encapsulated tumor cells is challenging because the arrangement of microspots of a matrix-cell droplet onto each well of a microwell plate is inconsistent and difficult to standardize. In addition, relatively low scales and lack of reproducibility discourage the application of three-dimensional organoid-based drug screening data for precision treatment or drug discovery. To overcome these limitations, we manufactured an automated organospotter-integrated high-throughput organo-on-pillar (high-TOP) drug-screening platform. Our system is compatible with various extracellular matrices, including BM extract, Matrigel, collagen, and hydrogel. In addition, it can be readily utilized for high-content analyses by simply exchanging the bottom plates without disrupting the domes. Our system demonstrated considerable robustness, consistency, reproducibility, and biological relevancy in three-dimensional drug sensitivity analyses using Matrigel-encapsulated ovarian cancer cell lines. We also demonstrated proof-of-concept cases representing the clinical feasibility of high-TOP-assisted ex vivo drug tests linked to clinical chemo-response in ovarian cancer patients. In conclusion, our platform provides an automated and standardized method for ex vivo drug-sensitivity-guided clinical response prediction, suggesting effective chemotherapy regimens for patients with cancer.
Collapse
Affiliation(s)
- Hye Ryeong Jun
- Central R & D Center, Medical & Bio Decision (MBD) Co., Ltd. Suwon, South Korea
| | - Hyun Ju Kang
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Sung Hun Ju
- Central R & D Center, Medical & Bio Decision (MBD) Co., Ltd. Suwon, South Korea
| | - Jung Eun Kim
- Central R & D Center, Medical & Bio Decision (MBD) Co., Ltd. Suwon, South Korea
| | - Sang Youl Jeon
- Central R & D Center, Medical & Bio Decision (MBD) Co., Ltd. Suwon, South Korea
| | - Bosung Ku
- Central R & D Center, Medical & Bio Decision (MBD) Co., Ltd. Suwon, South Korea
| | - Jae Jun Lee
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Minsung Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Min Jeong Kim
- Department of Obstetrics and Gynecology, Gynecologic Cancer Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jung-Joo Choi
- Department of Obstetrics and Gynecology, Gynecologic Cancer Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joseph J Noh
- Department of Obstetrics and Gynecology, Gynecologic Cancer Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hyun-Soo Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Gynecologic Cancer Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Jin-Ku Lee
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, South Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea; Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea.
| | - Dong Woo Lee
- Department of Biomedical Engineering, Gachon University, Seongnam, South Korea.
| |
Collapse
|
16
|
Tumor organoid biobank-new platform for medical research. Sci Rep 2023; 13:1819. [PMID: 36725963 PMCID: PMC9892604 DOI: 10.1038/s41598-023-29065-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/30/2023] [Indexed: 02/03/2023] Open
Abstract
Organoids are a new type of 3D model for tumor research, which makes up for the shortcomings of cell lines and xenograft models, and promotes the development of personalized precision medicine. Long-term culture, expansion and storage of organoids provide the necessary conditions for the establishment of biobanks. Biobanks standardize the collection and preservation of normal or pathological specimens, as well as related clinical information. The tumor organoid biobank has a good quality control system, which is conducive to the clinical transformation and large-scale application of tumor organoids, such as disease modeling, new drug development and high-throughput drug screening. This article summarized the common tumor types of patient-derived organoid (PDO) biobanks and the necessary information for biobank construction, such as the number of organoids, morphology, success rate of culture and resuscitation, pathological types. In our results, we found that patient-derived tumor organoid (PDTO) biobanks were being established more and more, with the Netherlands, the United States, and China establishing the most. Biobanks of colorectal, pancreas, breast, glioma, and bladder cancers were established more, which reflected the relative maturity of culture techniques for these tumors. In addition, we provided insights on the precautions and future development direction of PDTO biobank building.
Collapse
|
17
|
Qin T, Fan J, Lu F, Zhang L, Liu C, Xiong Q, Zhao Y, Chen G, Sun C. Harnessing preclinical models for the interrogation of ovarian cancer. J Exp Clin Cancer Res 2022; 41:277. [PMID: 36114548 PMCID: PMC9479310 DOI: 10.1186/s13046-022-02486-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/05/2022] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer (OC) is a heterogeneous malignancy with various etiology, histopathology, and biological feature. Despite accumulating understanding of OC in the post-genomic era, the preclinical knowledge still undergoes limited translation from bench to beside, and the prognosis of ovarian cancer has remained dismal over the past 30 years. Henceforth, reliable preclinical model systems are warranted to bridge the gap between laboratory experiments and clinical practice. In this review, we discuss the status quo of ovarian cancer preclinical models which includes conventional cell line models, patient-derived xenografts (PDXs), patient-derived organoids (PDOs), patient-derived explants (PDEs), and genetically engineered mouse models (GEMMs). Each model has its own strengths and drawbacks. We focus on the potentials and challenges of using these valuable tools, either alone or in combination, to interrogate critical issues with OC.
Collapse
|
18
|
Li Y, Wu J, Zhu G. Efficacy Analysis of Comprehensive Nursing in the Care of Ovarian Carcinoma Treated with Paclitaxel Combined with Nedaplatin. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9398823. [PMID: 36110573 PMCID: PMC9470341 DOI: 10.1155/2022/9398823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022]
Abstract
Objective To determine the effectiveness of comprehensive nursing in the care of ovarian carcinoma (OC) patients treated with paclitaxel (PTX) plus nedaplatin (NDP). Methods The research population comprised 180 advanced OC patients who received treatment in the Shaanxi Cancer Hospital between November 2018 and November 2021. The enrolled cases were assigned to two groups based on different nursing plans: an observation group (OG) with 100 cases treated with comprehensive nursing and a control group (CG) with 80 cases intervened by conventional nursing. Intergroup comparisons were performed to identify statistical significance in terms of the following parameters: serum NGF, TK1, and CA15-3 levels; VAS, SAS, and SDS scores; nursing compliance; incidence of adverse reactions; and nursing satisfaction. Results Compared with CG, OG showed the following: (1) lower posttreatment NGF, TK1, and CA15-3 levels; (2) lower scores of SAS and SDS; (3) higher nursing compliance; and (4) lower incidence of adverse reactions and higher nursing satisfaction after nursing. Conclusions Comprehensive nursing far outperformed conventional nursing in the care of advanced OC patients treated with PTX plus NDP, which is worth popularizing.
Collapse
Affiliation(s)
- Yahui Li
- Department of Gynecology and Oncology, Shaanxi Cancer Hospital, Xi'an, 710061 Shaanxi, China
| | - Jing Wu
- Department of General Surgery, Shaanxi Cancer Hospital, Xi'an, 710061 Shaanxi, China
| | - Gehong Zhu
- Department of Gynecology and Oncology, Shaanxi Cancer Hospital, Xi'an, 710061 Shaanxi, China
| |
Collapse
|
19
|
Liu Z, Wu J, Wang X, Ji X. Multivariate logistic regression analysis of the correlation between five biomarkers and ovarian cancer in patients with intermediate-risk: A prospective cross-sectional study. Front Cell Dev Biol 2022; 10:876071. [PMID: 36120557 PMCID: PMC9470860 DOI: 10.3389/fcell.2022.876071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/18/2022] [Indexed: 11/30/2022] Open
Abstract
Objective: To find potential diagnostic biomarkers for ovarian cancer (OC), a prospective analysis of the expression of five biomarkers in patients with intermediate-risk and their correlation with the occurrence of OC was conducted. Method: A prospective observational study was carried out, patients who underwent surgical treatment with benign or malignant ovarian tumors in our hospital from January 2020 to February 2021 were included in this study, and a total of 263 patients were enrolled. Based on the postoperative pathological results, enrolled patients were divided into ovarian cancer group and benign tumor group (n = 135). The ovarian cancer group was further divided into a mid-stage group (n = 46) and an advanced-stage group (n = 82). The basic information of the three groups of patients was collected, the preoperative imaging data of the patients were collected to assess the lymph node metastasis, the preoperative blood samples were collected to examine cancer antigen 125 (CA125), carbohydrate antigen 19–9 (CA19–9), Neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), and the postoperative pathological data were sorted and summarized. Result: The average during of disease in the advanced ovarian cancer group was 0.55 ± 0.18 years higher than the benign tumor group (0.43 ± 0.14 years), p < 0.001. In the advanced ovarian cancer group, the ratio of patients with the tumor, node, metastasis (TNM) stage IV (64.63%), with tumor Grade stage II and III (93.90%), and without lymph node metastasis (64.63%) was respectively more than that in the mid-stage group (accordingly 0.00, 36.96, 23.91%) (p < 0.001); The ratio of patients with TNM grade III in the mid-stage group (73.91%) was more than that in the advanced group (35.37%) (p < 0.001). The levels of the five biomarkers: CA19-9, CA125, NLR, PLR, and BDNF were different among the three groups (p < 0.001). Conclusion: CA19-9, CA125, NLR, PLR, BDNF are five biomarkers related to the occurrence of ovarian cancer and are risk factors for it. These five biomarkers and their Combined-Value may be suitable to apply in the diagnosis and the identification of ovarian cancer in patients with intermediate-risk.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Nuclear Medicine, Cangzhou Central Hospital, Cangzhou, China
| | - Jingjing Wu
- Department of Laboratory Medicine, Cangzhou Central Hospital, Cangzhou, China
| | - Xiuli Wang
- Department of Laboratory Medicine, Cangzhou Central Hospital, Cangzhou, China
| | - Xiaoyang Ji
- Department of Obstetrics and Gynecology, Ningjin Hospital of Integrated Traditional Chinese and Western Medicine, Xingtai, China
| |
Collapse
|
20
|
Narayan NJC, Requena D, Lalazar G, Ramos-Espiritu L, Ng D, Levin S, Shebl B, Wang R, Hammond WJ, Saltsman JA, Gehart H, Torbenson MS, Clevers H, LaQuaglia MP, Simon SM. Human liver organoids for disease modeling of fibrolamellar carcinoma. Stem Cell Reports 2022; 17:1874-1888. [PMID: 35803261 PMCID: PMC9391427 DOI: 10.1016/j.stemcr.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 11/29/2022] Open
Abstract
Fibrolamellar carcinoma (FLC) is a rare, often lethal, liver cancer affecting adolescents and young adults, for which there are no approved therapeutics. The development of therapeutics is hampered by a lack of in vitro models. Organoids have shown utility as a model system for studying many diseases. In this study, tumor tissue and the adjacent non-tumor liver were obtained at the time of surgery. The tissue was dissociated and grown as organoids. We developed 21 patient-derived organoid lines: 12 from metastases, three from the liver tumor and six from adjacent non-tumor liver. These patient-derived FLC organoids recapitulate the histologic morphology, immunohistochemistry, and transcriptome of the patient tumor. Patient-derived FLC organoids were used in a preliminary high-throughput drug screen to show proof of concept for the identification of therapeutics. This model system has the potential to improve our understanding of this rare cancer and holds significant promise for drug testing and development.
Collapse
Affiliation(s)
- Nicole J C Narayan
- Pediatric Surgical Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Laboratory of Cellular Biophysics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - David Requena
- Laboratory of Cellular Biophysics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Gadi Lalazar
- Laboratory of Cellular Biophysics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Lavoisier Ramos-Espiritu
- High Throughput and Spectroscopy Center, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Denise Ng
- Laboratory of Cellular Biophysics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Solomon Levin
- Laboratory of Cellular Biophysics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Bassem Shebl
- Laboratory of Cellular Biophysics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Ruisi Wang
- Laboratory of Cellular Biophysics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - William J Hammond
- Pediatric Surgical Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Laboratory of Cellular Biophysics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - James A Saltsman
- Pediatric Surgical Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Laboratory of Cellular Biophysics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Helmuth Gehart
- Hubrecht Institute, KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, the Netherlands
| | - Michael S Torbenson
- Department of Laboratory Medicine and Anatomic Pathology, Mayo Clinic, Rochester, MN, USA
| | - Hans Clevers
- Hubrecht Institute, KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, the Netherlands
| | - Michael P LaQuaglia
- Pediatric Surgical Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sanford M Simon
- Laboratory of Cellular Biophysics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
21
|
Yao W, Li S, Liu R, Jiang M, Gao L, Lu Y, Liang X, Zhang H. Long non-coding RNA PVT1: A promising chemotherapy and radiotherapy sensitizer. Front Oncol 2022; 12:959208. [PMID: 35965522 PMCID: PMC9373174 DOI: 10.3389/fonc.2022.959208] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/30/2022] [Indexed: 12/14/2022] Open
Abstract
The long non-coding RNA (lncRNA) PVT1 was first found to activate variant translocations in the plasmacytoma of mice. Human lncPVT1 is located on chromosome 8q24.21, at the same locus as the well-known MYC oncogene. LncPVT1 has been found to promote the progression of various malignancies. Chemoresistance and radioresistance seriously affect tumor treatment efficacy and are associated with the dysregulation of physiological processes in cancer cells, including apoptosis, autophagy, stemness (for cancer stem cells, CSC), hypoxia, epithelial–mesenchymal transition (EMT), and DNA damage repair. Previous studies have also implicated lncPVT1 in the regulation of these physiological mechanisms. In recent years, lncPVT1 was found to modulate chemoresistance and radioresistance in some cancers. In this review, we discuss the mechanisms of lncPVT1-mediated regulation of cellular chemoresistance and radioresistance. Due to its high expression in malignant tumors and sensitization effect in chemotherapy and radiotherapy, lncPVT1 is expected to become an effective antitumor target and chemotherapy and radiotherapy sensitizer, which requires further study.
Collapse
Affiliation(s)
- Weiping Yao
- Graduate Department, Bengbu Medical College, Bengbu, China
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Shuang Li
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Jinzhou Medical University, Jinzhou, China
| | - Ruiqi Liu
- Graduate Department, Bengbu Medical College, Bengbu, China
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Mingyun Jiang
- Graduate Department, Bengbu Medical College, Bengbu, China
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Liang Gao
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yanwei Lu
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xiaodong Liang
- Graduate Department, Bengbu Medical College, Bengbu, China
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Haibo Zhang, zhbdoctor @163.com; Xiaodong Liang,
| | - Haibo Zhang
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Haibo Zhang, zhbdoctor @163.com; Xiaodong Liang,
| |
Collapse
|
22
|
Li H, Liu H, Chen K. Living biobank-based cancer organoids: prospects and challenges in cancer research. Cancer Biol Med 2022; 19:j.issn.2095-3941.2021.0621. [PMID: 35856555 PMCID: PMC9334762 DOI: 10.20892/j.issn.2095-3941.2021.0621] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/28/2022] [Indexed: 11/24/2022] Open
Abstract
Biobanks bridge the gap between basic and translational research. Traditional cancer biobanks typically contain normal and tumor tissues, and matched blood. However, biospecimens in traditional biobanks are usually nonrenewable. In recent years, increased interest has focused on establishing living biobanks, including organoid biobanks, for the collection and storage of viable and functional tissues for long periods of time. The organoid model is based on a 3D in vitro cell culture system, is highly similar to primary tissues and organs in vivo, and can recapitulate the phenotypic and genetic characteristics of target organs. Publications on cancer organoids have recently increased, and many types of cancer organoids have been used for modeling cancer processes, as well as for drug discovery and screening. On the basis of the current research status, more exploration of cancer organoids through technical advancements is required to improve reproducibility and scalability. Moreover, given the natural characteristics of organoids, greater attention must be paid to ethical considerations. Here, we summarize recent advances in cancer organoid biobanking research, encompassing rectal, gastric, pancreatic, breast, and glioblastoma cancers. Living cancer biobanks that contain cancerous tissues and matched organoids with different genetic backgrounds, subtypes, and individualized characteristics will eventually contribute to the understanding of cancer and ultimately facilitate the development of innovative treatments.
Collapse
Affiliation(s)
- Haixin Li
- Cancer Biobank, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin 300060, China
| | - Hongkun Liu
- Cancer Biobank, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin 300060, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin 300060, China
| |
Collapse
|
23
|
Ou M, Li Q, Ling X, Yao J, Mo X. Cocktail Formula and Application Prospects for Oral and Maxillofacial Organoids. Tissue Eng Regen Med 2022; 19:913-925. [PMID: 35612711 DOI: 10.1007/s13770-022-00455-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 11/24/2022] Open
Abstract
Oral and maxillofacial organoids (OMOs), tiny tissues and organs derived from stem cells cultured through 3-d cell culture models, can fully summarize the cell tissue structure, physiological functions and biological characteristics of the source tissues in the body. OMOs are applied in areas such as disease modelling, developmental and regenerative medicine, drug screening, personalized treatment, etc. Although the construction of organoids in various parts of the oral and maxillofacial (OM) region has achieved considerable success, the existing cocktail formulae (construction strategies) are not widely applicable for tissues of various sources due to factors including the heterogeneity of the source tissues and the dependence on laboratory technology. Most of their formulae are based on growth factor niches containing expensive recombinant proteins with their efficiency remaining to be improved. In view of this, the cocktail formulae of various parts of the OM organs are reviewed with further discussion of the application and prospects for those OMOs to find some affordable cocktail formula with strong operability and high repeatability for various maxillofacial organs. The results may help improve the efficiency of organoid construction in the laboratory and accelerate the pace of the clinical use of organoid technology.
Collapse
Affiliation(s)
- Mingyu Ou
- Youjiang Medical University for Nationalities, No. 98 Countryside Road, BaiseGuangxi, 533000, China.,Department of Stomatology, China Affiliated Hospital of Youjiang Medical University for Nationalities, No. 18 Second Zhongshan Road, BaiseGuangxi, 533000, China
| | - Qing Li
- Youjiang Medical University for Nationalities, No. 98 Countryside Road, BaiseGuangxi, 533000, China.,Department of Stomatology, China Affiliated Hospital of Youjiang Medical University for Nationalities, No. 18 Second Zhongshan Road, BaiseGuangxi, 533000, China
| | - Xiaofang Ling
- Youjiang Medical University for Nationalities, No. 98 Countryside Road, BaiseGuangxi, 533000, China.,Department of Stomatology, China Affiliated Hospital of Youjiang Medical University for Nationalities, No. 18 Second Zhongshan Road, BaiseGuangxi, 533000, China
| | - Jinguang Yao
- Youjiang Medical University for Nationalities, No. 98 Countryside Road, BaiseGuangxi, 533000, China. .,Department of Stomatology, China Affiliated Hospital of Youjiang Medical University for Nationalities, No. 18 Second Zhongshan Road, BaiseGuangxi, 533000, China.
| | - Xiaoqiang Mo
- Youjiang Medical University for Nationalities, No. 98 Countryside Road, BaiseGuangxi, 533000, China.
| |
Collapse
|
24
|
Hicks WH, Bird CE, Gattie LC, Shami ME, Traylor JI, Shi DD, McBrayer SK, Abdullah KG. Creation and Development of Patient-Derived Organoids for Therapeutic Screening in Solid Cancer. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-022-00211-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
25
|
Cao C, Lan X, Shang B, Jiang W, Guo L, Zheng S, Bi X, Zhou A, Sun Z, Shou J. Phenotypical screening on metastatic PRCC-TFE3 fusion translocation renal cell carcinoma organoids reveals potential therapeutic agents. Clin Transl Oncol 2022; 24:1333-1346. [PMID: 35118587 PMCID: PMC9192364 DOI: 10.1007/s12094-021-02774-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/31/2021] [Indexed: 12/27/2022]
Abstract
PURPOSE Translocation renal cell carcinoma (tRCC) is a subtype that occurs predominantly in children and young individuals. Metastatic tRCC occurring in young patients is more aggressive than that occurring in older patients, and there are still no effective therapies. Organoids can mimic original tissues and be assessed by high-throughput screening (HTS). We aimed to utilize patient-derived organoids and HTS to screen drugs that can be repurposed for metastatic tRCC with PRCC-TFE3 fusion. METHODS Tumor tissues were obtained from treatment-naïve metastatic tRCC patients who underwent surgery. Histopathology and fluorescence in situ hybridization (FISH) confirmed the tRCC. Organoids derived from the dissected tissues were cultured and verified by FISH and RNA-seq. HTS was performed to seek promising drugs, and potential mechanisms were explored by RNA-seq and cell-based studies. RESULTS We successfully established a metastatic tRCC organoid with PRCC-TFE3 fusion, a common fusion subtype, and its characteristics were verified by histopathology, FISH, and RNA-seq. An HTS assay was developed, and the robustness was confirmed. A compound library of 1816 drugs was screened. Eventually, axitinib, crizotinib, and JQ-1 were selected for further validation and were found to induce cell cycle arrest and apoptosis. RNA-seq analyses of posttreatment organoids indicated that crizotinib induced significant changes in autophagy-related genes, consistent with the potential pathogenesis of tRCC. CONCLUSIONS We established and validated organoids derived from tissues dissected from a patient with metastatic tRCC with PRCC-TFE3 fusion and achieved the HTS process for the first time. Crizotinib might be a targeted therapy worthy of exploration in the clinic for metastatic tRCC with PRCC-TFE3 fusion. Such organoid and HTS assays may represent a promising model system in translational research assisting in the development of clinical strategies.
Collapse
Affiliation(s)
- Chuanzhen Cao
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17#, Chaoyang District, Beijing, 100021, People's Republic of China
| | - Xiaomei Lan
- K2 Oncology Co. Ltd., Beijing, 100176, People's Republic of China
| | - Bingqing Shang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17#, Chaoyang District, Beijing, 100021, People's Republic of China
| | - Weixing Jiang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17#, Chaoyang District, Beijing, 100021, People's Republic of China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Shan Zheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Xingang Bi
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17#, Chaoyang District, Beijing, 100021, People's Republic of China
| | - Aiping Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17#, Beijing, 100021, People's Republic of China.
| | - Zhijian Sun
- K2 Oncology Co. Ltd., Beijing, 100176, People's Republic of China.
| | - Jianzhong Shou
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17#, Chaoyang District, Beijing, 100021, People's Republic of China.
| |
Collapse
|
26
|
Mendoza-Martinez AK, Loessner D, Mata A, Azevedo HS. Modeling the Tumor Microenvironment of Ovarian Cancer: The Application of Self-Assembling Biomaterials. Cancers (Basel) 2021; 13:5745. [PMID: 34830897 PMCID: PMC8616551 DOI: 10.3390/cancers13225745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/07/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer (OvCa) is one of the leading causes of gynecologic malignancies. Despite treatment with surgery and chemotherapy, OvCa disseminates and recurs frequently, reducing the survival rate for patients. There is an urgent need to develop more effective treatment options for women diagnosed with OvCa. The tumor microenvironment (TME) is a key driver of disease progression, metastasis and resistance to treatment. For this reason, 3D models have been designed to represent this specific niche and allow more realistic cell behaviors compared to conventional 2D approaches. In particular, self-assembling peptides represent a promising biomaterial platform to study tumor biology. They form nanofiber networks that resemble the architecture of the extracellular matrix and can be designed to display mechanical properties and biochemical motifs representative of the TME. In this review, we highlight the properties and benefits of emerging 3D platforms used to model the ovarian TME. We also outline the challenges associated with using these 3D systems and provide suggestions for future studies and developments. We conclude that our understanding of OvCa and advances in materials science will progress the engineering of novel 3D approaches, which will enable the development of more effective therapies.
Collapse
Affiliation(s)
- Ana Karen Mendoza-Martinez
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK;
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Daniela Loessner
- Department of Chemical Engineering, Faculty of Engineering, Monash University, Melbourne, VIC 3800, Australia;
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Melbourne, VIC 3800, Australia
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia
- Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden e.V., 01069 Dresden, Germany
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK;
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Helena S. Azevedo
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK;
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| |
Collapse
|
27
|
Kou X, Ding H, Li L, Chao H. Caseinolytic protease P (CLPP) activated by ONC201 inhibits proliferation and promotes apoptosis in human epithelial ovarian cancer cells by inducing mitochondrial dysfunction. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1463. [PMID: 34734015 PMCID: PMC8506775 DOI: 10.21037/atm-21-4321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/09/2021] [Indexed: 12/23/2022]
Abstract
Background Caseinolytic protease P (CLPP) is a mitochondrial specific protein which has been reported to be related to tumor cell apoptosis. This study aims to explore the roles of CLPP in human epithelial ovarian cancer (EOC). Methods We determined CLPP expression in paracancerous tissues and cancer tissues obtained from 20 EOC patients, and also in 4 EOC cell lines, and one normal ovarian cell line (IOSE-80). We knocked CLPP expression down in SK-OV-3 and A2780 cells and overexpressed it in SW626 and OVcar3 cells. The effect of CLPP expression on cell proliferation, mitochondrial membrane potential, and apoptosis was then assessed by flow cytometry assay. Furthermore, the effect of ONC201 (agonist of CLPP) on the EOC cell lines was also investigated. Results The CLPP expression was markedly down-regulated in EOC cancer tissues, and the Kaplan-Meier Plotter database revealed its low expression was linked to poor prognosis in EOC patients. Low expression of CLPP up-regulated the expression of NADH: ubiquinone oxidoreductase subunit A12 (NDUFA12), succinate dehydrogenase complex flavoprotein subunit A (SDHA), and succinate dehydrogenase complex iron sulfur subunit B (SDHB), which are key members of the mitochondrial respiratory chain, and these up-regulated proteins further led to the increase of mitochondrial membrane potential, cell proliferation promotion and neoplasm metastasis. Conversely, while overexpression of CLPP led to the opposite results, including inducing the decrease of mitochondrial membrane potential and apoptosis. In addition, stimulation with ONC201 enhanced the function of CLPP in SW626 and OVcar3 cells, and silencing of CLPP could neutralize the effect of ONC201. Conclusions Our findings suggest that CLPP mediated mitochondrial dysfunction inhibits the proliferation and migration of EOC cells.
Collapse
Affiliation(s)
- Xinxin Kou
- Department of Gynaecology, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Hui Ding
- Department of Gynaecology, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Lei Li
- Department of Gynaecology, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Hongtu Chao
- Department of Gynaecology, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
28
|
Chen H, Liu Y, Liu P, Dai Q, Wang P. LINC01094 promotes the invasion of ovarian cancer cells and regulates the Wnt/β-catenin signaling pathway by targeting miR-532-3p. Exp Ther Med 2021; 22:1228. [PMID: 34539824 PMCID: PMC8438678 DOI: 10.3892/etm.2021.10662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 06/04/2021] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) participate in the development of ovarian cancer (OC). The present study aimed to explore the roles of long intergenic non-protein coding RNA 1094 (LINC01094) in OC. LINC01094 and microRNA (miR)-532-3p expression in OC tissues and cells were measured using reverse transcription-quantitative PCR. Cell migration and invasion were detected using wound healing assays and Transwell assays, respectively. The binding of LINC01094 or β-catenin to miR-126-5p was detected using a Dual-luciferase reporter assay, and protein expression was confirmed using western blot analysis. The expression level of LINC01094 in patients with OC was higher in OC tissues compared with in adjacent tissues, and LINC01094 was upregulated in OC cell lines. In addition, LINC01094 overexpression promoted the viability, migration, invasion and cell cycle progression of OC cells, and inhibited OC cell apoptosis. Moreover, LINC01094 negatively regulated miR-532-3p in OC cells and tissues. miR-532-3p overexpression decreased the viability, migration, invasion and cell cycle progression of OC cells alongside downregulation of Wnt/β-catenin signaling pathway protein expression, as well as increasing OC cell apoptosis. Inhibition of LINC01094 with small interfering (si)-LINC01094 and overexpression of LINC01094 respectively reversed the effect of miR-532-3p inhibitor and mimics on OC cells. miR-532-3p could directly target β-catenin, and miR-532-3p inhibitor increased β-catenin expression, while si-LINC01094 attenuated this effect. In addition, LINC01094 overexpression promoted tumor growth in vivo by regulating miR-532-3p. Taken together, LINC01094 promoted the growth, migration, invasion and Wnt/β-catenin signaling pathway expression of OC cells by modulating miR-532-3p.
Collapse
Affiliation(s)
- Haiyan Chen
- Department of Gynaecology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| | - Yanlin Liu
- Department of Gynaecology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| | - Ping Liu
- Department of Reproductive Medicine, Hainan West Central Hospital, Danzhou, Hainan 571799, P.R. China
| | - Qiuxiang Dai
- Department of Obstetrical and Gynecology, Hainan Modern Women and Children's Hospital, Haikou, Hainan 570300, P.R. China
| | - Peiliang Wang
- Department of Gynaecology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| |
Collapse
|
29
|
Pozzi S, Scomparin A, Israeli Dangoor S, Rodriguez Ajamil D, Ofek P, Neufeld L, Krivitsky A, Vaskovich-Koubi D, Kleiner R, Dey P, Koshrovski-Michael S, Reisman N, Satchi-Fainaro R. Meet me halfway: Are in vitro 3D cancer models on the way to replace in vivo models for nanomedicine development? Adv Drug Deliv Rev 2021; 175:113760. [PMID: 33838208 DOI: 10.1016/j.addr.2021.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/24/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022]
Abstract
The complexity and diversity of the biochemical processes that occur during tumorigenesis and metastasis are frequently over-simplified in the traditional in vitro cell cultures. Two-dimensional cultures limit researchers' experimental observations and frequently give rise to misleading and contradictory results. Therefore, in order to overcome the limitations of in vitro studies and bridge the translational gap to in vivo applications, 3D models of cancer were developed in the last decades. The three dimensions of the tumor, including its cellular and extracellular microenvironment, are recreated by combining co-cultures of cancer and stromal cells in 3D hydrogel-based growth factors-inclusive scaffolds. More complex 3D cultures, containing functional blood vasculature, can integrate in the system external stimuli (e.g. oxygen and nutrient deprivation, cytokines, growth factors) along with drugs, or other therapeutic compounds. In this scenario, cell signaling pathways, metastatic cascade steps, cell differentiation and self-renewal, tumor-microenvironment interactions, and precision and personalized medicine, are among the wide range of biological applications that can be studied. Here, we discuss a broad variety of strategies exploited by scientists to create in vitro 3D cancer models that resemble as much as possible the biology and patho-physiology of in vivo tumors and predict faithfully the treatment outcome.
Collapse
Affiliation(s)
- Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy
| | - Sahar Israeli Dangoor
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniel Rodriguez Ajamil
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Paula Ofek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lena Neufeld
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniella Vaskovich-Koubi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Pradip Dey
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shani Koshrovski-Michael
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Noa Reisman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
30
|
Roles of Mesenchymal Stem Cell-Derived Exosomes in Cancer Development and Targeted Therapy. Stem Cells Int 2021; 2021:9962194. [PMID: 34335792 PMCID: PMC8289580 DOI: 10.1155/2021/9962194] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/25/2021] [Indexed: 01/02/2023] Open
Abstract
Exosomes have emerged as a new drug delivery system. In particular, exosomes derived from mesenchymal stem cells (MSCs) have been extensively studied because of their tumor-homing ability and yield advantages. Considering that MSC-derived exosomes are a double-edged sword in the development, metastasis, and invasion of tumors, engineered exosomes have broad potential use. In this review, we focused on the latest development in the treatment of tumors using engineered and nonengineered MSC-derived exosomes (MSC-EXs). Nonengineered MSC-EXs exert an antitumor effect on several well-studied tumors by affecting tumor growth, angiogenesis, metastasis, and invasion. Furthermore, engineered exosomes have promising research prospects as drug-carrying tools for the transport of miRNAs, small-molecule drugs, and proteins. Although exosomes lack uniform standards in terms of definition, separation, and purification, they still have great research value because of their unique advantages, such as high biocompatibility and low toxicity. Future studies on MSC-EXs should elucidate the mechanisms underlying their anticancer effect and the safety of their application.
Collapse
|
31
|
Chang X, Dong Y. CACNA1C is a prognostic predictor for patients with ovarian cancer. J Ovarian Res 2021; 14:88. [PMID: 34210324 PMCID: PMC8252246 DOI: 10.1186/s13048-021-00830-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/28/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND CACNA1C, as a type of voltage-dependent calcium ion transmembrane channel, played regulatory roles in the development and progress of multiple tumors. This study was aimed to analyze the roles of CACNA1C in ovarian cancer (OC) of overall survival (OS) and to explore its relationships with immunity. METHODS Single gene mRNA sequencing data and corresponding clinical information were obtained from The Cancer Genome Atlas Database (TCGA) and the International Cancer Genome Consortium (ICGC) datasets. Gene set enrichment analysis (GSEA) was used to identify CACNA1C-related signal pathways. Univariate and multivariate Cox regression analyses were applied to evaluate independent prognostic factors. Besides, associations between CACNA1C and immunity were also explored. RESULTS CACNA1C had a lower expression in OC tumor tissues than in normal tissues (P < 0.001), with significant OS (P = 0.013) and a low diagnostic efficiency. We further validated the expression levels of CACNA1C in OC by means of the ICGC dataset (P = 0.01), qRT-PCR results (P < 0.001) and the HPA database. Univariate and multivariate Cox hazard regression analyses indicated that CACNA1C could be an independent risk factor of OS for OC patients (both P < 0.001). Five significant CACNA1C-related signaling pathways were identified by means of GSEA. As for genetic alteration analysis, altered CACNA1C groups were significantly associated with OS (P = 0.0169), progression-free survival (P = 0.0404), disease-free survival (P = 0.0417) and disease-specific survival (P = 9.280e-3), compared with unaltered groups in OC. Besides, CACNA1C was dramatically associated with microsatellite instability (MSI) and immunity. CONCLUSIONS Our results shed light on that CACNA1C could be a prognostic predictor of OS in OC and it was closely related to immunity.
Collapse
Affiliation(s)
- Xiaohan Chang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Liaoning Province, 110004, Shenyang, P.R. China
| | - Yunxia Dong
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Liaoning Province, 110004, Shenyang, P.R. China.
| |
Collapse
|
32
|
Zeng L, Liao Q, Zhao H, Jiang S, Yang X, Tang H, He Q, Yang X, Fang S, He J, Cui W, Huang L, Ma S, Cui S. Raltitrexed as a synergistic hyperthermia chemotherapy drug screened in patient-derived colorectal cancer organoids. Cancer Biol Med 2021; 18:j.issn.2095-3941.2020.0566. [PMID: 33710819 PMCID: PMC8330527 DOI: 10.20892/j.issn.2095-3941.2020.0566] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/26/2020] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Organoids have recently been used as in vitro models to screen chemotherapy drugs in combination with hyperthermia treatment in colorectal cancer. Our research aimed to establish a library of patient-derived colorectal cancer organoids to evaluate synergism between chemotherapy drugs and hyperthermia; validate an index of the hyperthermia chemotherapy sensitization enhancement ratio (HCSER) to identify the chemotherapeutics most enhanced by hyperthermia; and recommend chemotherapy drugs for hyperthermic intraperitoneal treatment. METHODS Organoids were grown from cells extracted from colorectal cancer patient samples or colorectal cancer cell lines. Cells from both sources were encapsulated in 3D Matrigel droplets, which were formulated in microfluidics and phase-transferred into identical cell-laden Matrigel microspheres. The microspheres were seeded in 96-well plates, with each well containing a single microsphere that developed into an organoid after 7 days. The organoids were used to evaluate the efficacy of chemotherapy drugs at both 37°C as a control and 43°C for 90 min to examine hyperthermia synergism. Cell viability was counted with 10% CCK8. RESULTS We successfully established a library of colorectal cancer organoids from 22 patient parental tumors. We examined the hyperthermia synergism of 7 commonly used hyperthermic intraperitoneal chemotherapy drugs. In 11 of the 22 patient organoids, raltitrexed had significant hyperthermia synergism, which was indexed as the highest HCSER score within each patient group. CONCLUSIONS Our results primarily demonstrated the use of patient-derived colorectal cancer organoids as in vitro models to evaluate hyperthermia synergistic chemotherapeutics. We found that hyperthermia enhanced the effect of raltitrexed the most among the common anti-colorectal cancer drugs.
Collapse
Affiliation(s)
- Lisi Zeng
- Institute of Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Quanxing Liao
- Department of Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Haoran Zhao
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
| | - Shengwei Jiang
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Gene and Antibody Therapy, Tsinghua University, Shenzhen 518055, China
| | - Xianzi Yang
- Department of Medical Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Hongsheng Tang
- Department of Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Qingjun He
- Department of Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Xiansheng Yang
- Department of Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Shuxian Fang
- Department of Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Jinfu He
- Department of Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Weiwen Cui
- Department of Bioengineering, University of California, Berkeley 94720, USA
| | - Laiqiang Huang
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Gene and Antibody Therapy, Tsinghua University, Shenzhen 518055, China
| | - Shaohua Ma
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
| | - Shuzhong Cui
- Department of Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| |
Collapse
|
33
|
Yang X, Xu X, Zhu H, Wang M, Wang D. Organoid research in digestive system tumors. Oncol Lett 2021; 21:308. [PMID: 33732384 PMCID: PMC7905586 DOI: 10.3892/ol.2021.12569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/04/2021] [Indexed: 12/13/2022] Open
Abstract
Digestive system tumors are the most common cause of cancer-associated mortality worldwide, although their underlying biological behavior still requires further investigation. Most of the in vitro studies that have been published have been based on the two-dimensional (2D) culture system. However, digestive system tumors exhibit considerable histological and functional heterogeneity, and clonal diversity and heterogeneity cannot be entirely reflected in the 2D culture system. Recently, the development of organoids appears to have shed some light on this area of cancer research. The present review discusses the recent advancements that have been made in the development of several specific organoids in digestive system solid tumors.
Collapse
Affiliation(s)
- Xiaoxiao Yang
- Department of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212031, P.R. China
| | - Xuewen Xu
- Department of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212031, P.R. China
| | - Haitao Zhu
- Department of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212031, P.R. China
| | - Ming Wang
- Department of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212031, P.R. China
| | - Dongqing Wang
- Department of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212031, P.R. China
| |
Collapse
|
34
|
Li M, Zhao L, Zhou X, Zhang K, Yin P, Liu S, Zou Y, Li Q. Detection of carcinoma in serous effusions: a review. Am J Cancer Res 2021; 11:43-60. [PMID: 33520359 PMCID: PMC7840719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/20/2020] [Indexed: 06/12/2023] Open
Abstract
A malignant serous effusion is one of the most common complications of advanced tumors, indicating a poor prognosis and having a profound impact on diagnosis, treatment, and prognosis. It is of great significance to identify benign and malignant effusions quickly and accurately. Both cellular and non-cellular components in the effusion can be employed for detection, diagnostic methods are necessary to obtain a definite diagnosis and more relevant information such as tumor classification. In this review, we focus on the comparison of several widespread cytological preparation methods, enrichment technology of exfoliated cells, and present tests for serous effusions, mainly including routine and special stains, immunocytochemistry, electron microscopy, enzyme-linked immunosorbent assay, flow cytometry, and molecular analysis.
Collapse
Affiliation(s)
- Min Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, China
| | - Lanbo Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, China
| | - Xue Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, China
| | - Kailu Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, China
| | - Panyue Yin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, China
| | - Shuhua Liu
- College of Chemistry Engineering, Tianjin UniversityTianjin 300350, China
| | - Yuliang Zou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, China
| | - Qiling Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, China
| |
Collapse
|
35
|
Ding DC, Chang YH, Wu KC, Harnod T. The organoid: A research model for ovarian cancer. Tzu Chi Med J 2021; 34:255-260. [PMID: 35912056 PMCID: PMC9333109 DOI: 10.4103/tcmj.tcmj_63_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 11/29/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is a heterogeneous disease with a variety of distinct clinical and molecular characteristics. The currently available and common research models for EOC include tumor cell lines and patient-derived xenografts. However, these models have certain shortcomings: establishing a cell line is time-consuming, loss of genetic traits after long-term culture is a possibility, and investment is required in terms of animal care facilities. Therefore, better research models are required. Organoid technology was originally developed from colorectal cancer. Tumor organoid is a three-dimensional culture system and can help accurately recapture the tumor phenotype from the original tumor. Tumor organoid systems can overcome the above-mentioned shortcomings of the currently available research models. The organoid model can be used for culturing ovarian cancer subtypes, screening drugs, assessing genomes, and establishing biobanks. However, the currently available organoid models can only culture one type of cells, epithelial cells. Therefore, an organoid-on-a-chip device can be developed in the future to provide a microenvironment for cell–cell, cell–matrix, and cell–media interactions. Thus, organoid models can be used in ovarian cancer research and can generate a simulated in vivo system, enabling studies on the heterogeneity of ovarian cancer.
Collapse
|
36
|
Sheta R, Bachvarova M, Plante M, Renaud MC, Sebastianelli A, Gregoire J, Navarro JM, Perez RB, Masson JY, Bachvarov D. Development of a 3D functional assay and identification of biomarkers, predictive for response of high-grade serous ovarian cancer (HGSOC) patients to poly-ADP ribose polymerase inhibitors (PARPis): targeted therapy. J Transl Med 2020; 18:439. [PMID: 33213473 PMCID: PMC7678187 DOI: 10.1186/s12967-020-02613-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 11/06/2020] [Indexed: 11/13/2022] Open
Abstract
Background Poly(ADP-ribose) polymerase inhibitors (PARPis) specifically target homologous recombination deficiency (HRD) cells and display good therapeutic effect in women with advanced-stage BRCA1/2-mutated breast and epithelial ovarian cancer (EOC). However, about 50% of high grade serous ovarian cancers (HGSOC) present with HRD due to epigenetic BRCA1 inactivation, as well as genetic/epigenetic inactivation(s) of other HR genes, a feature known as “BRCAness”. Therefore, there is a potential for extending the use of PARPis to these patients if HR status can be identified. Methods We have developed a 3D (spheroid) functional assay to assess the sensitivity of two PARPis (niraparib and olaparib) in ascites-derived primary cell cultures (AsPCs) from HGSOC patients. A method for AsPCs preparation was established based on a matrix (agarose), allowing for easy isolation and successive propagation of monolayer and 3D AsPCs. Based on this method, we performed cytotoxicity assays on 42 AsPCs grown both as monolayers and spheroids. Results The response to PARPis treatment in monolayer AsPCs, was significantly higher, compared to 3D AsPCs, as 88% and 52% of the monolayer AsPCs displayed sensitivity to niraparib and olaparib respectively, while 66% of the 3D AsPCs were sensitive to niraparib and 38% to olaparib, the latter being more consistent with previous estimates of HRD (40%–60%) in EOC. Moreover, niraparib displayed a significantly stronger cytotoxic effect in both in 3D and monolayer AsPCs, which was confirmed by consecutive analyses of the HR pathway activity (γH2AX foci formation) in PARPis-sensitive and resistant AsPCs. Global gene expression comparison of 6 PARPi-resistant and 6 PARPi-sensitive 3D AsPCs was indicative for the predominant downregulation of numerous genes and networks with previously demonstrated roles in EOC chemoresistance, suggesting that the PARPis-sensitive AsPCs could display enhanced sensitivity to other chemotherapeutic drugs, commonly applied in cancer management. Microarray data validation identified 24 potential gene biomarkers associated with PARPis sensitivity. The differential expression of 7 selected biomarkers was consecutively confirmed by immunohistochemistry in matched EOC tumor samples. Conclusion The application of this assay and the potential biomarkers with possible predictive significance to PARPis therapy of EOC patients now need testing in the setting of a clinical trial.
Collapse
Affiliation(s)
- Razan Sheta
- Department of Molecular Medicine, Université Laval, Québec, QC, G1V 0A6, Canada.,Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada
| | - Magdalena Bachvarova
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada
| | - Marie Plante
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Marie-Claude Renaud
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Alexandra Sebastianelli
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Jean Gregoire
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Jamilet Miranda Navarro
- Bioinformatics Department, Center for Genetic Engineering and Biotechnology, 10600, Havana, CP, Cuba
| | - Ricardo Bringas Perez
- Bioinformatics Department, Center for Genetic Engineering and Biotechnology, 10600, Havana, CP, Cuba
| | - Jean-Yves Masson
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Molecular Biology, Medical Biochemistry, and Pathology, Laval University Cancer Research Center, Québec, QC, G1V 0A6, Canada
| | - Dimcho Bachvarov
- Department of Molecular Medicine, Université Laval, Québec, QC, G1V 0A6, Canada. .,Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.
| |
Collapse
|
37
|
Yang C, Xia BR, Zhang ZC, Zhang YJ, Lou G, Jin WL. Immunotherapy for Ovarian Cancer: Adjuvant, Combination, and Neoadjuvant. Front Immunol 2020; 11:577869. [PMID: 33123161 PMCID: PMC7572849 DOI: 10.3389/fimmu.2020.577869] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy. Surgery and chemotherapy are the primary treatments for ovarian cancer; however, patients often succumb to recurrence with chemotherapeutic resistance within several years after the initial treatment. In the past two decades, immunotherapy has rapidly developed, and has revolutionized the treatment of various types of cancer. Despite the fact that immunotherapy response rates among ovarian cancer patients remain modest, treatment with immune checkpoint inhibitors (ICIs), chimeric antigen receptor (CAR)- and TCR-engineered T cells is rapidly developing. Therapeutic efficiency could be improved significantly if immunotherapy is included as an adjuvant therapy, in combination with chemotherapy, radiation therapy, and the use of anti-angiogenesis drugs, and poly ADP ribose polymerase inhibitors (PARPi). Newly developed technologies that identify therapeutic targets, predict treatment efficacy, rapidly screen potential immunotherapy drugs, provide neoadjuvant immunotherapy, and utilize nanomedicine technology provide new opportunities for the treatment of ovarian cancer, and have the potential to prolong patient survival. However, important issues that may hinder the efficacy of such approaches, including hyperprogressive disease (HPD), immunotherapy-resistance, and toxicity of the treatments, including neurotoxicity, must be taken into account and addressed for these therapies to be effective.
Collapse
Affiliation(s)
- Chang Yang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Bai-Rong Xia
- Department of Gynecology Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhao-Cong Zhang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yong-Jian Zhang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ge Lou
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wei-Lin Jin
- Department of Instrument Science and Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
38
|
Gralewska P, Gajek A, Marczak A, Rogalska A. Participation of the ATR/CHK1 pathway in replicative stress targeted therapy of high-grade ovarian cancer. J Hematol Oncol 2020; 13:39. [PMID: 32316968 PMCID: PMC7175546 DOI: 10.1186/s13045-020-00874-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/08/2020] [Indexed: 12/15/2022] Open
Abstract
Ovarian cancer is one of the most lethal gynecologic malignancies reported throughout the world. The initial, standard-of-care, adjuvant chemotherapy in epithelial ovarian cancer is usually a platinum drug, such as cisplatin or carboplatin, combined with a taxane. However, despite surgical removal of the tumor and initial high response rates to first-line chemotherapy, around 80% of women will develop cancer recurrence. Effective strategies, including chemotherapy and new research models, are necessary to improve the prognosis. The replication stress response (RSR) is characteristic of the development of tumors, including ovarian cancer. Hence, RSR pathway and DNA repair proteins have emerged as a new area for anticancer drug development. Although clinical trials have shown poly (ADP-ribose) polymerase inhibitors (PARPi) response rates of around 40% in women who carry a mutation in the BRCA1/2 genes, PARPi is responsible for tumor suppression, but not for complete tumor regression. Recent reports suggest that cells with impaired homologous recombination (HR) activities due to mutations in TP53 gene or specific DNA repair proteins are specifically sensitive to ataxia telangiectasia and Rad3-related protein (ATR) inhibitors. Replication stress activates DNA repair checkpoint proteins (ATR, CHK1), which prevent further DNA damage. This review describes the use of DNA repair checkpoint inhibitors as single agents and strategies combining these inhibitors with DNA-damaging compounds for ovarian cancer therapy, as well as the new platforms used for optimizing ovarian cancer therapy.
Collapse
Affiliation(s)
- Patrycja Gralewska
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, Institute of Biophysics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Arkadiusz Gajek
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, Institute of Biophysics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Agnieszka Marczak
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, Institute of Biophysics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Aneta Rogalska
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, Institute of Biophysics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland.
| |
Collapse
|