1
|
De Cock L, Palubeckaitė I, Bersani F, Faehling T, Pasquali S, Umbaugh S, Meister MT, Danks MR, Manasterski P, Miallot R, Krumbholz M, Roohani S, Heymann D, Cidre-Aranaz F, Wozniak A, Schöffski P, Bovée JVMG, Merlini A, Venneker S. Establishment of patient-derived 3D in vitro models of sarcomas: literature review and guidelines on behalf of the FORTRESS working group. Neoplasia 2025; 65:101171. [PMID: 40324303 DOI: 10.1016/j.neo.2025.101171] [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: 03/21/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Sarcomas are a large family of rare and heterogeneous mesenchymal tumors, which respond poorly to available systemic treatments. Translation of preclinical findings into clinical applications has been slow, limiting improvements in patients' outcomes and ultimately highlighting the need for a better understanding of sarcoma biology to develop more effective, subtype-specific therapies. To this end, reliable preclinical models are crucial, but the development of 3D in vitro sarcoma models has been lagging behind that of epithelial cancers. This is primarily due to the rarity and heterogeneity of sarcomas, and lack of widespread knowledge regarding the optimal growth conditions of these in vitro models. In this review, we provide an overview of currently available sarcoma tumoroid models, together with guidelines and suggestions for model development and characterization, on behalf of the FORTRESS (Forum For Translational Research in Sarcomas) international research working group on 3D sarcoma models.
Collapse
Affiliation(s)
- Lore De Cock
- Laboratory of Experimental Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium; Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Ieva Palubeckaitė
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Francesca Bersani
- Department of Oncology, Translational Oncology Laboratory "Paola Gilardi", University of Turin, Turin, Italy
| | - Tobias Faehling
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Sandro Pasquali
- Molecular Pharmacology, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sam Umbaugh
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Division of Applied Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Torsten Meister
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Molly R Danks
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Piotr Manasterski
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard Miallot
- Department of Surgical and Interventional Sciences, McGill University, Montreal, QC, Canada; Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Manuela Krumbholz
- University Hospital Erlangen, Department of Pediatrics Erlangen, Germany
| | - Siyer Roohani
- Charité - Universitätsmedizin Berlin, corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) Clinician Scientist Program, Berlin, Germany
| | - Dominique Heymann
- Nantes Université, CNRS, UMR6286, US2B, Institut de Cancérologie de l'Ouest, Saint-Herblain, France; Université of Sheffield, School of Medicine and Population Health, Sheffield, United Kingdom
| | - Florencia Cidre-Aranaz
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Agnieszka Wozniak
- Laboratory of Experimental Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Patrick Schöffski
- Laboratory of Experimental Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium; Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Judith V M G Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Alessandra Merlini
- Department of Oncology, Translational Oncology Laboratory "Paola Gilardi", University of Turin, Turin, Italy; Division of Medical Oncology, San Luigi Gonzaga University Hospital, Orbassano, Turin, Italy
| | - Sanne Venneker
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
2
|
Hiraga H, Machida R, Kawai A, Kunisada T, Yonemoto T, Endo M, Nishida Y, Nagano A, Ae K, Yoshida S, Asanuma K, Toguchida J, Furuta T, Nakayama R, Akisue T, Hiruma T, Morii T, Nishimura H, Hiraoka K, Takeyama M, Emori M, Tsukushi S, Hatano H, Kawashima H, Isu K, Tanaka K, Kataoka T, Fukuda H, Iwamoto Y, Ozaki T. Methotrexate, Doxorubicin, and Cisplatin Versus Methotrexate, Doxorubicin, and Cisplatin + Ifosfamide in Poor Responders to Preoperative Chemotherapy for Newly Diagnosed High-Grade Osteosarcoma (JCOG0905): A Multicenter, Open-Label, Randomized Trial. J Clin Oncol 2025:JCO2401281. [PMID: 40138604 DOI: 10.1200/jco-24-01281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 01/02/2025] [Accepted: 01/24/2025] [Indexed: 03/29/2025] Open
Abstract
PURPOSE Our previous NECO phase II studies on high-grade osteosarcoma suggested that administering ifosfamide (IF; 16 g/m2 [4g/m2 once on day 1, then 2g/m2 once on days 2-7] × six) to patients showing a poor response (PrRsp) to preoperative chemotherapy with methotrexate, doxorubicin, and cisplatin (MAP) improves their prognoses. In this Japan Clinical Oncology Group (JCOG) study, JCOG0905, we aimed to investigate the efficacy and safety of IF in patients with PrRsp. METHODS JCOG0905 is a multicenter, open-label, multi-institutional, randomized trial. Eligible patients (50 years and younger) had resectable, high-grade osteosarcoma (stage II or III, Union for International Cancer Control TNM) of the extremities, limb girdles, and thoracic wall. After two MAP cycles and tumor resection, patients with PrRsp were randomly assigned to either the MAP or MAP plus 15 g/m2 (3g/m2 once daily on days 1-5) × six IF (MAP + IF [MAPIF]) group. The primary end point was disease-free survival (DFS); secondary end points were overall survival (OS) and safety. The planned sample size was 100 patients with a one-sided α of .1 and a power of 0.7, assuming a 3-year DFS of 50% and 65% for MAP and MAPIF, respectively. This trial is registered with the Japan Registry of Clinical Trials (jRCT; jRCTs031180126). RESULTS Of the 287 patients registered between February 2010 and August 2020, 51 and 52 patients with PrRsp were assigned to the MAP and MAPIF groups, respectively. As of March 2022, DFS did not differ between groups (hazard ratio [HR], 1.05 [95% CI, 0.55 to 1.98]) and OS was numerically inferior in the MAPIF group (HR, 1.48 [95% CI, 0.68 to 3.22]). Nine and zero patients in the MAPIF and MAP groups discontinued treatment because of adverse events, respectively. CONCLUSION Evidence from JCOG0905 does not support the addition of IF for patients with PrRsp.
Collapse
Affiliation(s)
- Hiroaki Hiraga
- Musculoskeletal Oncology, NHO Hokkaido Cancer Center, Sapporo, Japan
| | - Ryunosuke Machida
- Japan Clinical Oncology Group Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Akira Kawai
- Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Toshiyuki Kunisada
- Orthopaedic Surgery, Faculty of Medicine, Okayama University, Okayama, Japan
| | | | - Makoto Endo
- Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Nishida
- Department of Rehabilitation Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Akihito Nagano
- Orthopaedic Surgery, Gifu University School of Medicine, Gifu, Japan
| | - Keisuke Ae
- Orthopaedic Surgery, Cancer Institute Hospital, Tokyo, Japan
| | - Shinichirou Yoshida
- Orthopaedic Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Junya Toguchida
- Department of Tissue Regeneration, Center for Induced Pluripotent Stem Cell Research and Application, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Taisuke Furuta
- Orthopaedic Surgery, Hiroshima University, Hiroshima, Japan
| | - Robert Nakayama
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | | | - Toru Hiruma
- Bone and Soft Tissue Tumour Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Takeshi Morii
- Orthopaedic Surgery, Kyorin University, Tokyo, Japan
| | - Hideki Nishimura
- Orthopaedic Surgery, Kagawa University Hospital, Takamatsu, Japan
| | - Koji Hiraoka
- Orthopaedic Surgery, Kurume University Hospital, Kurume, Japan
| | - Masanobu Takeyama
- Orthopaedic Surgery, Yokohama City University Hospital, Yokohama, Japan
| | - Makoto Emori
- Orthopaedic Surgery, Sapporo Medical University Hospital, Sapporo, Japan
| | | | - Hiroshi Hatano
- Musculoskeletal Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Hiroyuki Kawashima
- Orthopaedic Surgery, Niigata University Medical & Dental Hospital, Niigata, Japan
| | - Kazuo Isu
- Orthopaedic Surgery, Higashi Sapporo Hospital, Sapporo, Japan
| | - Kazuhiro Tanaka
- Advanced Medical Sciences, Oita University Faculty of Medicine, Oita, Japan
| | - Tomoko Kataoka
- Japan Clinical Oncology Group Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Haruhiko Fukuda
- Japan Clinical Oncology Group Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | | | - Toshifumi Ozaki
- Orthopaedic Surgery, Faculty of Medicine, Okayama University, Okayama, Japan
| |
Collapse
|
3
|
Costa S, Rodrigues J, Vieira C, Dias S, Viegas J, Castro F, Sarmento B, Leite Pereira C. Advancing osteosarcoma 3D modeling in vitro for novel tumor microenvironment-targeted therapies development. J Control Release 2024; 376:1068-1085. [PMID: 39505219 DOI: 10.1016/j.jconrel.2024.10.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024]
Abstract
Osteosarcoma (OS) represents one of the most common primary bone cancers affecting children and young adults. The available treatments have remained unimproved for the past decades, hampered by the poor knowledge of OS etiology/pathophysiology and the lack of innovative, predictive and biologically relevant in vitro models, that can recapitulate the 3D OS tumor microenvironment (TME). Here, we report the development and characterization of an innovative 3D model of OS, composed of OS tumor cells, immune cells (macrophages) and mesenchymal stem cells (MSCs), that formed a multicellular tissue spheroid (MCTS). This fully humanized 3D model was shown to accurately mimic the native histological features of OS, while innately leading to the polarization of macrophages towards an M2-like phenotype, highly aggressive and pro-tumor profile. Upon the exposure to immunomodulatory molecules, the MCTS were shown to be responsive by shifting macrophages polarization, and dramatically altering the TME secretome. In agreement, when treated with immunomodulatory/stimulatory nanoparticles (NPSs), we were able to revert the TME secretome towards an anti-inflammatory profile. This study establishes an advanced 3D OS model capable of shedding light on macrophages and MSCs contributions to disease progression, paving the way for the development of innovative therapeutic approaches targeting the OS TME, while providing a biologically relevant in vitro tool for the efficacy screening of novel OS therapeutic approaches.
Collapse
Affiliation(s)
- Sofia Costa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - João Rodrigues
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Carolina Vieira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; FMUP - Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Sofia Dias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Juliana Viegas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Catarina Leite Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.
| |
Collapse
|
4
|
Ye HS, Zhou D, Li H, Lv J, Huang HQ, She JJ, Nie JH, Li TT, Lu MD, Du BL, Yang SQ, Chen PX, Li S, Ye GL, Luo W, Liu J. Organoid forming potential as complementary parameter for accurate evaluation of breast cancer neoadjuvant therapeutic efficacy. Br J Cancer 2024; 130:1109-1118. [PMID: 38341511 PMCID: PMC10991527 DOI: 10.1038/s41416-024-02595-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND 13-15% of breast cancer/BC patients diagnosed as pathological complete response/pCR after neoadjuvant systemic therapy/NST suffer from recurrence. This study aims to estimate the rationality of organoid forming potential/OFP for more accurate evaluation of NST efficacy. METHODS OFPs of post-NST residual disease/RD were checked and compared with clinical approaches to estimate the recurrence risk. The phenotypes of organoids were classified via HE staining and ER, PR, HER2, Ki67 and CD133 immuno-labeling. The active growing organoids were subjected to drug sensitivity tests. RESULTS Of 62 post-NST BC specimens, 24 were classified as OFP-I with long-term active organoid growth, 19 as OFP-II with stable organoid growth within 3 weeks, and 19 as OFP-III without organoid formation. Residual tumors were overall correlated with OFP grades (P < 0.001), while 3 of the 18 patients (16.67%) pathologically diagnosed as tumor-free (ypT0N0M0) showed tumor derived-organoid formation. The disease-free survival/DFS of OFP-I cases was worse than other two groups (Log-rank P < 0.05). Organoids of OFP-I/-II groups well maintained the biological features of their parental tumors and were resistant to the drugs used in NST. CONCLUSIONS The OFP would be a complementary parameter to improve the evaluation accuracy of NST efficacy of breast cancers.
Collapse
Affiliation(s)
- Hai-Shan Ye
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Dan Zhou
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, China
| | - Hong Li
- Biomedical Laboratory, Guangzhou Jingke BioTech Group, Guangzhou, 510005, China
| | - Jin Lv
- Department of Pathology, The First People's Hospital of Foshan, Foshan, 528100, China
| | - Hui-Qi Huang
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, China
| | - Jia-Jun She
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, 528100, China
| | - Jun-Hua Nie
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Ting-Ting Li
- Biomedical Laboratory, Guangzhou Jingke BioTech Group, Guangzhou, 510005, China
| | - Meng-Di Lu
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Bo-Le Du
- Biomedical Laboratory, Guangzhou Jingke BioTech Group, Guangzhou, 510005, China
| | - Shu-Qing Yang
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, China
| | - Pei-Xian Chen
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, China
| | - Sheng Li
- Biomedical Laboratory, Guangzhou Jingke BioTech Group, Guangzhou, 510005, China
| | - Guo-Lin Ye
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528100, China.
| | - Wei Luo
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, 528100, China.
| | - Jia Liu
- School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Liaoning Laboratory of Cancer Genetics and Epigenetics, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
5
|
White B, Swietach P. What can we learn about acid-base transporters in cancer from studying somatic mutations in their genes? Pflugers Arch 2024; 476:673-688. [PMID: 37999800 PMCID: PMC11006749 DOI: 10.1007/s00424-023-02876-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 11/25/2023]
Abstract
Acidosis is a chemical signature of the tumour microenvironment that challenges intracellular pH homeostasis. The orchestrated activity of acid-base transporters of the solute-linked carrier (SLC) family is critical for removing the end-products of fermentative metabolism (lactate/H+) and maintaining a favourably alkaline cytoplasm. Given the critical role of pH homeostasis in enabling cellular activities, mutations in relevant SLC genes may impact the oncogenic process, emerging as negatively or positively selected, or as driver or passenger mutations. To address this, we performed a pan-cancer analysis of The Cancer Genome Atlas simple nucleotide variation data for acid/base-transporting SLCs (ABT-SLCs). Somatic mutation patterns of monocarboxylate transporters (MCTs) were consistent with their proposed essentiality in facilitating lactate/H+ efflux. Among all cancers, tumours of uterine corpus endometrial cancer carried more ABT-SLC somatic mutations than expected from median tumour mutation burden. Among these, somatic mutations in SLC4A3 had features consistent with meaningful consequences on cellular fitness. Definitive evidence for ABT-SLCs as 'cancer essential' or 'driver genes' will have to consider microenvironmental context in genomic sequencing because bulk approaches are insensitive to pH heterogeneity within tumours. Moreover, genomic analyses must be validated with phenotypic outcomes (i.e. SLC-carried flux) to appreciate the opportunities for targeting acid-base transport in cancers.
Collapse
Affiliation(s)
- Bobby White
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK.
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| |
Collapse
|
6
|
Xu R, Chen R, Tu C, Gong X, Liu Z, Mei L, Ren X, Li Z. 3D Models of Sarcomas: The Next-generation Tool for Personalized Medicine. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:171-186. [PMID: 38884054 PMCID: PMC11169319 DOI: 10.1007/s43657-023-00111-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/18/2024]
Abstract
Sarcoma is a complex and heterogeneous cancer that has been difficult to study in vitro. While two-dimensional (2D) cell cultures and mouse models have been the dominant research tools, three-dimensional (3D) culture systems such as organoids have emerged as promising alternatives. In this review, we discuss recent developments in sarcoma organoid culture, with a focus on their potential as tools for drug screening and biobanking. We also highlight the ways in which sarcoma organoids have been used to investigate the mechanisms of gene regulation, drug resistance, metastasis, and immune interactions. Sarcoma organoids have shown to retain characteristics of in vivo biology within an in vitro system, making them a more representative model for sarcoma research. Our review suggests that sarcoma organoids offer a potential path forward for translational research in this field and may provide a platform for developing personalized therapies for sarcoma patients.
Collapse
Affiliation(s)
- Ruiling Xu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| | - Ruiqi Chen
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| | - Xiaofeng Gong
- College of Life Science, Fudan University, Shanghai, 200433 China
| | - Zhongyue Liu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| | - Lin Mei
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| | - Xiaolei Ren
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha, 410011 Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, No. 139 Renmin Road, Changsha, 410011 Hunan China
| |
Collapse
|
7
|
Krajnović M, Kožik B, Božović A, Jovanović-Ćupić S. Multiple Roles of the RUNX Gene Family in Hepatocellular Carcinoma and Their Potential Clinical Implications. Cells 2023; 12:2303. [PMID: 37759525 PMCID: PMC10527445 DOI: 10.3390/cells12182303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most frequent cancers in humans, characterised by a high resistance to conventional chemotherapy, late diagnosis, and a high mortality rate. It is necessary to elucidate the molecular mechanisms involved in hepatocarcinogenesis to improve diagnosis and treatment outcomes. The Runt-related (RUNX) family of transcription factors (RUNX1, RUNX2, and RUNX3) participates in cardinal biological processes and plays paramount roles in the pathogenesis of numerous human malignancies. Their role is often controversial as they can act as oncogenes or tumour suppressors and depends on cellular context. Evidence shows that deregulated RUNX genes may be involved in hepatocarcinogenesis from the earliest to the latest stages. In this review, we summarise the topical evidence on the roles of RUNX gene family members in HCC. We discuss their possible application as non-invasive molecular markers for early diagnosis, prognosis, and development of novel treatment strategies in HCC patients.
Collapse
Affiliation(s)
| | - Bojana Kožik
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovića Alasa 12-14, Vinča, 11351 Belgrade, Serbia; (M.K.); (A.B.); (S.J.-Ć.)
| | | | | |
Collapse
|
8
|
Lin TC. RUNX2 and Cancer. Int J Mol Sci 2023; 24:ijms24087001. [PMID: 37108164 PMCID: PMC10139076 DOI: 10.3390/ijms24087001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/03/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Runt-related transcription factor 2 (RUNX2) is critical for the modulation of chondrocyte osteoblast differentiation and hypertrophy. Recently discovered RUNX2 somatic mutations, expressional signatures of RUNX2 in normal tissues and tumors, and the prognostic and clinical significance of RUNX2 in many types of cancer have attracted attention and led RUNX2 to be considered a biomarker for cancer. Many discoveries have illustrated the indirect and direct biological functions of RUNX2 in orchestrating cancer stemness, cancer metastasis, angiogenesis, proliferation, and chemoresistance to anticancer compounds, warranting further exploration of the associated mechanisms to support the development of a novel therapeutic strategy. In this review, we focus mainly on critical and recent research developments, including RUNX2's oncogenic activities, by summarizing and integrating the findings on somatic mutations of RUNX2, transcriptomic studies, clinical information, and discoveries about how the RUNX2-induced signaling pathway modulates malignant progression in cancer. We also comprehensively discuss RUNX2 RNA expression in a pancancer panel and in specific normal cell types at the single-cell level to indicate the potential cell types and sites for tumorigenesis. We expect this review to shed light on the recent mechanistical findings and modulatory role of RUNX2 in cancer progression and provide biological information that can guide new research in this field.
Collapse
Affiliation(s)
- Tsung-Chieh Lin
- Genomic Medicine Core Laboratory, Department of Medical Research and Development, Chang Gung Memorial Hospital, Linkou 333, Taiwan
- Department of Biomedical Sciences, Chang Gung University, Taoyuan City 333, Taiwan
| |
Collapse
|
9
|
Auxenochlorella pyrenoidosa extract supplementation replacing fetal bovine serum for Carassius auratus muscle cell culture under low-serum conditions. Food Res Int 2023; 164:112438. [PMID: 36738005 DOI: 10.1016/j.foodres.2022.112438] [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: 10/09/2022] [Revised: 12/25/2022] [Accepted: 12/29/2022] [Indexed: 01/02/2023]
Abstract
Cultured meat production requires large-scale cell proliferation in vitro with the supplementation of necessary media especially serum. This study investigated the capacity of Auxenochlorella pyrenoidosa extract (APE) to replace fetal bovine serum (FBS) for cell culture under low-serum conditions using Carassius auratus muscle (CAM) cells. Supplementation with APE and 5% FBS in the culture media significantly promoted the proliferation of CAM cells and increased the expression of MyoD in cells compared to that with 5% FBS through cell counting kit-8 and immunofluorescence staining assay. In addition, CAM cells in the media containing 5% FBS and APE could be continually cultured for 4 passages, and the cell number was 1.58 times higher than the counterpart without APE in long-term culture. Moreover, supplementation with APE realized large-scale culture on microcarriers under low-serum conditions, and more adherent cells were observed on microcarriers in 2% FBS supplemented with APE, compared with those in 2% FBS and 10% FBS without APE. These findings highlighted a potentially promising application of APE in muscle cell culture under low-serum conditions for cultured meat production.
Collapse
|
10
|
Dual Inhibition of BRAF-MAPK and STAT3 Signaling Pathways in Resveratrol-Suppressed Anaplastic Thyroid Cancer Cells with BRAF Mutations. Int J Mol Sci 2022; 23:ijms232214385. [PMID: 36430869 PMCID: PMC9692422 DOI: 10.3390/ijms232214385] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Anaplastic thyroid cancer is an extremely lethal malignancy without reliable treatment. BRAFV600E point mutation is common in ATCs, which leads to MAPK signaling activation and is regarded as a therapeutic target. Resveratrol inhibits ATC cell growth, while its impact on BRAF-MAPK signaling remains unknown. This study aims to address this issue by elucidating the statuses of BRAF-MAPK and STAT3 signaling activities in resveratrol-treated THJ-11T, THJ-16T, and THJ-21T ATC cells and Nthyori 3-1 thyroid epithelial cells. RT-PCR and Sanger sequencing revealed MKRN1-BRAF fusion mutation in THJ-16T, BRAF V600E point mutation in THJ-21T, and wild-type BRAF genes in THJ-11T and Nthyori 3-1 cells. Western blotting and immunocytochemical staining showed elevated pBRAF, pMEK, and pERK levels in THJ-16T and THJ-21T, but not in THJ-11T or Nthyori 3-1 cells. Calcein/PI, EdU, and TUNEL assays showed that compared with docetaxel and doxorubicin and MAPK-targeting dabrafenib and trametinib, resveratrol exerted more powerful inhibitory effects on mutant BRAF-harboring THJ-16T and THJ-21T cells, accompanied by reduced levels of MAPK pathway-associated proteins and pSTAT3. Trametinib- and dabrafenib-enhanced STAT3 activation was efficiently suppressed by resveratrol. In conclusion, resveratrol acts as dual BRAF-MAPK and STAT3 signaling inhibitor and a promising agent against ATCs with BRAF mutation.
Collapse
|
11
|
Nie JH, Yang T, Li H, Li S, Li TT, Ye HS, Lu MD, Chu X, Zhong GQ, Zhou JL, Wu ML, Zhang Y, Liu J. Frequently Expressed Glypican-3 As A Promising Novel Therapeutic Target for Osteosarcomas. Cancer Sci 2022; 113:3618-3632. [PMID: 35946078 PMCID: PMC9530858 DOI: 10.1111/cas.15521] [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: 05/04/2022] [Revised: 07/07/2022] [Accepted: 07/25/2022] [Indexed: 11/28/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone malignancy without a reliable therapeutic target. Glypican-3 (GPC3) mutation and upregulation have been detected in multi-drug resistant OS, and anti-GPC3 immunotherapy can effectively suppress the growth of organoids. Further profiling of GPC3 mutations and expression patterns in OS is of clinical significance. To address these issues, fresh OS specimens were collected from 24 patients for cancer-targeted next-generation sequencing (NGS) and three-dimensional patient-derived organoid (PDO) culture. A tumor microarray was prepared using 37 archived OS specimens. Immunohistochemical (IHC) staining was performed on OS specimens and microarrays to profile GPC3 and CD133 expression as well as intratumoral distribution patterns. RT-PCR was conducted to semi-quantify GPC3 and CD133 expression levels in the OS tissues. Anti-GPC3 immunotherapy was performed on OS organoids with or without GPC3 expression and its efficacy was analyzed using multiple experimental approaches. No OS cases with GPC3 mutations were found, except for the positive control (OS-08). IHC staining revealed GPC3 expression in 73.77% (45/61) of OSs in weak (+; 29/45), moderate (++; 8/45), and strong (+++; 8/45) immunolabeling densities. The intratumoral distribution of GPC3-positive cells was variable in the focal (+; 10-30%; 8/45), partial (++; 31-70%; 22/45), and the most positive patterns (+++; > 71%; 15/45), which coincided with CD133 immunolabeling (P = 9.89×10-10 ). The anti-GPC3 antibody efficiently inhibits Wnt/β-catenin signaling and induces apoptosis in GPC3-positive PDOs and PDXs, as opposed to GPC3-negative PDOs and PDXs. The high frequency of GPC3 and CD133 co-expression and the effectiveness of anti-wildtype GPC3-ab therapy in GPC3-positive OS models suggest that GPC3 is a novel prognostic parameter and a promising therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Jun-Hua Nie
- South China University of Technology School of Medicine, Guangzhou, China
| | - Tao Yang
- Department of Orthopedic Oncology, Guangdong Provincial People's Hospital Affiliated to South China University of Technology School of Medicine, Guangzhou, China
| | - Hong Li
- BioMed Laboratory, Guangzhou Jingke Biotech Group, Guangzhou, China
| | - Sheng Li
- BioMed Laboratory, Guangzhou Jingke Biotech Group, Guangzhou, China
| | - Ting-Ting Li
- BioMed Laboratory, Guangzhou Jingke Biotech Group, Guangzhou, China
| | - Hai-Shan Ye
- South China University of Technology School of Medicine, Guangzhou, China
| | - Meng-Di Lu
- South China University of Technology School of Medicine, Guangzhou, China
| | - Xiao Chu
- Department of Orthopedic Oncology, Guangdong Provincial People's Hospital Affiliated to South China University of Technology School of Medicine, Guangzhou, China
| | - Guo-Qing Zhong
- Department of Orthopedic Oncology, Guangdong Provincial People's Hospital Affiliated to South China University of Technology School of Medicine, Guangzhou, China
| | - Jie-Long Zhou
- Department of Orthopedic Oncology, Guangdong Provincial People's Hospital Affiliated to South China University of Technology School of Medicine, Guangzhou, China
| | - Mo-Li Wu
- Liaoning Laboratory of Cancer Genomics and Epigenomics, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yu Zhang
- Department of Orthopedic Oncology, Guangdong Provincial People's Hospital Affiliated to South China University of Technology School of Medicine, Guangzhou, China
| | - Jia Liu
- South China University of Technology School of Medicine, Guangzhou, China.,Liaoning Laboratory of Cancer Genomics and Epigenomics, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|