1
|
Ueda Y, Kiyonaka S, Selfors LM, Inoue K, Harada H, Doura T, Onuma K, Uchiyama M, Kurogi R, Yamada Y, Sun JH, Sakaguchi R, Tado Y, Omatsu H, Suzuki H, Aoun M, Nakayama T, Kajimoto T, Yano T, Holmdahl R, Hamachi I, Inoue M, Mori Y, Takahashi N. Intratumour oxidative hotspots provide a niche for cancer cell dissemination. Nat Cell Biol 2025; 27:530-543. [PMID: 39984655 DOI: 10.1038/s41556-025-01617-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/10/2025] [Indexed: 02/23/2025]
Abstract
Intratumour heterogeneity represents the hierarchical integration of genetic, phenotypic and microenvironmental heterogeneity. Although single-cell sequencing has clarified genetic and phenotypic variability, the heterogeneity of nongenetic, microenvironmental factors remains elusive. Here, we developed T-AP1, a tumour-targeted probe tracking extracellular H2O2, which allows the visualization and characterization of tumour cells exposed to oxidative stress, a hallmark of cancer. T-AP1 identified actively budding intratumour regions as H2O2-rich microenvironments (H2O2 hotspots), which were primarily established by neutrophils. Mechanistically, tumour cells exposed to H2O2 underwent partial epithelial-mesenchymal transition through p38-MYC axis activation and migrated away from H2O2 hotspots. This escape mechanism was absent in normal epithelial cells but prevalent in most cancers except NRF2-hyperactivated tumours, which exhibited abrogated p38 responses and enhanced antioxidant programmes, thus revealing an intrinsic stress defence programme in cancers. Together, T-AP1 enabled the identification of H2O2 hotspots that provide a niche for cancer cell dissemination, offering insights into metastasis initiation.
Collapse
Affiliation(s)
- Yoshifumi Ueda
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Shigeki Kiyonaka
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan.
- Department of Biomolecular Engineering, Nagoya University, Nagoya, Japan.
- Research Institute for Quantum and Chemical Innovation, Nagoya University, Nagoya, Japan.
| | - Laura M Selfors
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Keisuke Inoue
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Tomohiro Doura
- Department of Biomolecular Engineering, Nagoya University, Nagoya, Japan
| | - Kunishige Onuma
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Makoto Uchiyama
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Ryuhei Kurogi
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Yuji Yamada
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Jiacheng H Sun
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Reiko Sakaguchi
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Yuki Tado
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Haruki Omatsu
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Harufumi Suzuki
- Department of Biomolecular Engineering, Nagoya University, Nagoya, Japan
| | - Mike Aoun
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | - Takahiro Nakayama
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Taketoshi Kajimoto
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | | | - Rikard Holmdahl
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan.
| | - Nobuaki Takahashi
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan.
- The Hakubi Center for Advanced Research, Kyoto University, Kyoto, Japan.
| |
Collapse
|
2
|
Song X, Pan Z, Zhang Y, Yang W, Zhang T, Wang H, Chen Y, Yu X, Ding H, Li R, Ge P, Xu L, Dong G, Jiang F. Excessive MYC Orchestrates Macrophages induced Chromatin Remodeling to Sustain Micropapillary-Patterned Malignancy in Lung Adenocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2403851. [PMID: 39899538 PMCID: PMC11948069 DOI: 10.1002/advs.202403851] [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: 04/12/2024] [Revised: 01/15/2025] [Indexed: 02/05/2025]
Abstract
Current understanding of micropapillary (MP)-subtype lung adenocarcinoma (LUAD) remains confined to biological activities and genomic landscapes. Unraveling the major regulatory programs underlying MP patterned malignancy offers opportunities to identify more feasible therapeutic targets for patients with MP LUAD. This study shows that patients with MP subtype LUAD have aberrant activation of the MYC pathway compared to patients with other subtypes. In vitro and xenograft mouse model studies reveal that MP pattern in malignancy cannot be solely due to aberrant MYC expression but requires the involvement of M2-like macrophages. Excessively expressed MYC leads to the accumulation of M2-like macrophages from the bone marrow, which secretes TGFβ, to induce the expression of FOSL2 in tumor cells, thereby remodeling chromatin accessibility at promoter regions of MP-pattern genes to promote the MYC-mediated de novo transcriptional regulation of these genes. Additionally, the MP-pattern in malignancy can be effectively alleviated by disrupting the TGFβ-FOSL2 axis. These findings reveal new functions for the M2-like macrophage-TGFβ-FOSL2 axis in MYC-overexpressing MP-subtype LUAD, identifying targetable vulnerabilities in this pathway.
Collapse
Affiliation(s)
- Xuming Song
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Zehao Pan
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Yi Zhang
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Department of PathologyNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
| | - Wenmin Yang
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Department of PathologyNanjing Drum Tower hospitalNanjing210008P.R. China
| | - Te Zhang
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Department of Biochemistry and Molecular GeneticsFeinberg School of MedicineNorthwestern UniversityChicagoIllinois60201USA
| | - Hui Wang
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Yuzhong Chen
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Xinnian Yu
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Hanlin Ding
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Rutao Li
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Department of Thoracic SurgeryThe Fourth Affiliated Hospital of Soochow UniversityNanjing215000P. R. China
| | - Pengfei Ge
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Lin Xu
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Collaborative Innovation Center for Cancer Personalized MedicineNanjing Medical UniversityNanjing211116P. R. China
| | - Gaochao Dong
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
| | - Feng Jiang
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
| |
Collapse
|
3
|
Matsuura Y, Onuma K, Coppo R, Uematsu H, Kondo J, Miyagawa‐Hayashino A, Takeda‐Miyata N, Kameyama K, Furuya T, Okada S, Shimomura M, Inoue M, Inoue M. Dynamic change of polarity in spread through air spaces of pulmonary malignancies. J Pathol 2025; 265:260-273. [PMID: 39804150 PMCID: PMC11794978 DOI: 10.1002/path.6382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/16/2024] [Accepted: 11/22/2024] [Indexed: 02/06/2025]
Abstract
Spread through air spaces (STAS) is a histological finding of lung tumours where tumour cells exist within the air space of the lung parenchyma beyond the margin of the main tumour. Although STAS is an important prognostic factor, the pathobiology of STAS remains unclear. Here, we investigated the mechanism of STAS by analysing the relationship between STAS and polarity switching in vivo and in vitro. Histopathological analysis revealed that apical membranes were observed outside the STAS lesions around colorectal cancer (CRC) lung metastases and lung adenocarcinomas. When apical-out CRC organoids were administered intratracheally to mice, the organoids had greater metastatic potential than did single cells. To investigate the pathobiology of STAS, we established an in vitro model of STAS in which CRC or lung cancer organoids were co-cultured with 2D-cultured mouse airway epithelial organoids (2D-MAOs). Adhesion of cancer organoids to 2D-MAOs was much less than to type I collagen or endothelial cells, suggesting a protective role of the airway epithelium against adhesion. Loss of the apical membrane of CRC organoids at the contact surface with 2D-MAOs after adhesion was responsible for establishing adhesion. When airway epithelium was stimulated by transforming growth factor beta 1 (TGF-β1), adhesion of CRC organoids was enhanced. Among TGF-β1-induced genes in airway epithelium, follistatin-like protein 1 (FSTL1) increased CRC organoid adhesion by promoting loss of the apical membrane. These results suggested that TGF-β1-induced FSTL1 may promote metastatic progression of STAS by altering the polarity status. Elucidating the mechanism of STAS could contribute to the improvement of survival in patients with pulmonary malignancies associated with STAS. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Grants
- 21cm0106203h0006 Japan Agency for Medical Research and Development
- 18H02648 Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology in Japan
- 20H03772 Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology in Japan
- 20K08286 Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology in Japan
- 22K08982 Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology in Japan
- 23K07395 Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology in Japan
- Japan Agency for Medical Research and Development
Collapse
Affiliation(s)
- Yoshiaki Matsuura
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
- Divison of Thoracic Surgery, Department of Surgery, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Kunishige Onuma
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Roberto Coppo
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Hiroyuki Uematsu
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
- KBBM Inc.KyotoJapan
| | - Jumpei Kondo
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
- Department of Molecular Biology and Clinical Investigation, Graduate School of MedicineOsaka UniversityOsakaJapan
| | | | - Naoko Takeda‐Miyata
- Department of Surgical PathologyKyoto Prefectural University of MedicineKyotoJapan
| | - Kenji Kameyama
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
- Divison of Thoracic Surgery, Department of Surgery, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Tatsuo Furuya
- Divison of Thoracic Surgery, Department of Surgery, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Satoru Okada
- Divison of Thoracic Surgery, Department of Surgery, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Masanori Shimomura
- Divison of Thoracic Surgery, Department of Surgery, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Masayoshi Inoue
- Divison of Thoracic Surgery, Department of Surgery, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Masahiro Inoue
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
| |
Collapse
|
4
|
Li S, Gao S, Qin L, Ding C, Qu J, Cui Y, Qiang L, Yin S, Zheng X, Meng H. Micropapillary structure: A natural tumor collective invasion model with enhanced stem-like properties. Cancer Sci 2025; 116:308-321. [PMID: 39568148 PMCID: PMC11786311 DOI: 10.1111/cas.16396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/22/2024] Open
Abstract
Cancer stem cells aggregate to form clusters, which have enhanced stem-like properties and metastasis potential. However, the molecular mechanisms underlying the formation of cancer stem cell cluster-like structures with acquisition of stronger invasion and metastasis abilities remain unclear. Micropapillary carcinoma (MPC) is a subpopulation of small, merulioid, inverted, nonfibrous vascular clusters floating in the stroma present in a range of solid malignant tumors and characterized by frequent vascular/lymphatic vessel invasion and lymph node metastasis. Our results showed that these cell clusters exhibit a stem cell phenotype, supporting the premise that MPC may serve as a promising solid tumor model for studying invasion and metastasis of cancer stem cell clusters. In this review, we discuss the latest advances in MPC research and targeted therapy, focusing on analysis of their stem-like characteristics, mapping their multiomics characteristics, and elucidating the vascular and immune microenvironment of MPC. The existing MPC organoid model was employed to explore potential breakthroughs in targeted therapy and immunotherapy for cancer stem cell clusters.
Collapse
Affiliation(s)
- Sisi Li
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Shuangshu Gao
- Department of PathologyHarbin Medical UniversityHarbinChina
| | - Ling Qin
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Caixia Ding
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Jinghui Qu
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Yifei Cui
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Lixia Qiang
- Department of Respiratory MedicineThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Shengjie Yin
- Department of Medical OncologyMunicipal Hospital of ChifengChifengChina
| | - Xiaoyu Zheng
- Department of AnesthesiologyHarbin Medical University Cancer HospitalHarbinChina
| | - Hongxue Meng
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| |
Collapse
|
5
|
Durymanov M. Tumor Spheroids, Tumor Organoids, Tumor Explants, and Tumoroids: What Are the Differences between Them? BIOCHEMISTRY. BIOKHIMIIA 2025; 90:200-213. [PMID: 40254399 DOI: 10.1134/s0006297924604234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 04/22/2025]
Abstract
Three-dimensional (3D) cell cultures that mimic tumor microenvironment have become an essential tool in cancer research and drug response analysis, significantly enhancing our understanding of tumor biology and advancing personalized medicine. Currently, the most widely mentioned 3D multicellular culture models include spheroids, organoids, tumor explants, and tumoroids. These 3D structures, exploited for various applications, are generated from cancer and non-cancer cells of different origin using multiple techniques. However, despite extensive research and numerous studies, consistent definitions of these 3D culture models are not clearly established. The manuscript provides a comprehensive overview of these models, detailing brief history of their research, unique biological characteristics, advantages, limitations, and specific applications.
Collapse
Affiliation(s)
- Mikhail Durymanov
- Medical Informatics Laboratory, Yaroslav-the-Wise Novgorod State University, Veliky Novgorod, 173003, Russia.
- Department of Radiochemistry, Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
6
|
Paul CD, Yankaskas C, Shahi Thakuri P, Balhouse B, Salen S, Bullock A, Beam S, Chatman A, Djikeng S, Yang XJ, Wong G, Dey I, Holmes S, Dockey A, Bailey-Steinitz L, Zheng L, Li W, Chandra V, Nguyen J, Sharp J, Willems E, Kennedy M, Dallas MR, Kuninger D. Long-term maintenance of patient-specific characteristics in tumoroids from six cancer indications. Sci Rep 2025; 15:3933. [PMID: 39890889 PMCID: PMC11785764 DOI: 10.1038/s41598-025-86979-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025] Open
Abstract
Tumoroids, sometimes referred to as cancer organoids, are patient-derived cancer cells grown as 3D, self-organized multicellular structures that maintain key characteristics (e.g., genotype, gene expression levels) of the tumor from which they originated. These models have emerged as valuable tools for studying tumor biology, cytotoxicity, and response of patient-derived cells to cancer therapies. However, the establishment and maintenance of tumoroids has historically been challenging, labor intensive, and highly variable from lab to lab, hindering their widespread use. Here, we characterize the establishment and/or expansion of colorectal, lung, head and neck, breast, pancreas, and endometrial tumoroids using the standardized, serum-free Gibco OncoPro Tumoroid Culture Medium. Newly derived tumoroid lines (n = 20) were analyzed by targeted genomic profiling and RNA sequencing and were representative of tumor tissue samples. Tumoroid lines were stable for over 250 days in culture and freeze-thaw competent. Previously established tumoroid lines were also transitioned to OncoPro medium and exhibited, on average, similar growth rates and conserved donor-specific characteristics when compared to original media systems. Additionally, OncoPro medium was compatible with both embedded culture in extracellular matrix and growth in a suspension format for facile culture and scale up. An example application of these models for assessing the cytotoxicity of a natural killer cell line and primary natural killer cells over time and at various doses demonstrated the compatibility of these models with assays used in compound and cell therapy development. We anticipate that the standardization and versatility of this approach will have important benefits for basic cancer research, drug discovery, and personalized medicine and help make tumoroid models more accessible to the cancer research community.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sylvia Beam
- Thermo Fisher Scientific, Frederick, MD, USA
| | | | | | | | | | - Isha Dey
- Thermo Fisher Scientific, Bengaluru, Karnataka, India
| | | | | | | | - Lina Zheng
- Thermo Fisher Scientific, Carlsbad, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Nakagawa R, Beardsley A, Durney S, Hayward MK, Subramanyam V, Meyer NP, Wismer H, Goodarzi H, Weaver VM, Van de Mark D, Goga A. Tumor Cell Spatial Organization Directs EGFR/RAS/RAF Pathway Primary Therapy Resistance through YAP Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.26.615226. [PMID: 39386679 PMCID: PMC11463411 DOI: 10.1101/2024.09.26.615226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Non-small cell lung cancers (NSCLC) harboring common mutations in EGFR and KRAS characteristically respond transiently to targeted therapies against those mutations, but invariably, tumors recur and progress. Resistance often emerges through mutations in the therapeutic target or activation of alternative signaling pathways. Mechanisms of acute tumor cell resistance to initial EGFR (EGFRi) or KRASG12C (G12Ci) pathway inhibition remain poorly understood. Our study reveals that acute response to EGFR/RAS/RAF-pathway inhibition is spatial and culture context specific. In vivo, EGFR mutant tumor xenografts shrink by > 90% following acute EGFRi therapy, and residual tumor cells are associated with dense stroma and have increased nuclear YAP. Interestingly, in vitro EGFRi induced cell cycle arrest in NSCLC cells grown in monolayer, while 3D spheroids preferentially die upon inhibitor treatment. We find differential YAP nuclear localization and activity, driven by the distinct culture conditions, as a common resistance mechanism for selective EGFR/KRAS/BRAF pathway therapies. Forced expression of the YAPS127A mutant partially protects cells from EGFR-mediated cell death in spheroid culture. These studies identify YAP activation in monolayer culture as a non-genetic mechanism of acute EGFR/KRAS/BRAF therapy resistance, highlighting that monolayer vs spheroid cell culture systems can model distinct stages of patient cancer progression.
Collapse
Affiliation(s)
- Rachel Nakagawa
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Andrew Beardsley
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
- Department Of Medicine, University of California, San Francisco, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Sophia Durney
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Mary-Kate Hayward
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California San Francisco, San Francisco, CA, USA
| | - Vishvak Subramanyam
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
| | - Nathaniel P. Meyer
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Harrison Wismer
- Biological Imaging Development CoLab, UCSF, San Francisco, CA, USA
| | - Hani Goodarzi
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Valerie M Weaver
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California San Francisco, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Daniel Van de Mark
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Andrei Goga
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
- Department Of Medicine, University of California, San Francisco, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| |
Collapse
|
8
|
Grigoreva TA, Kindt DN, Sagaidak AV, Novikova DS, Tribulovich VG. Cellular Systems for Colorectal Stem Cancer Cell Research. Cells 2025; 14:170. [PMID: 39936962 PMCID: PMC11817814 DOI: 10.3390/cells14030170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Oncological diseases consistently occupy leading positions among the most life-threatening diseases, including in highly developed countries. At the same time, the second most common cause of cancer death is colorectal cancer. The current level of research shows that the development of effective therapy, in this case, requires a new grade of understanding processes during the emergence and development of a tumor. In particular, the concept of cancer stem cells that ensure the survival of chemoresistant cells capable of giving rise to new tumors is becoming widespread. To provide adequate conditions that reproduce natural processes typical for tumor development, approaches based on increasingly complex cellular systems are being improved. This review discusses the main strategies that allow for the study of the properties of tumor cells with an emphasis on colorectal cancer stem cells. The features of working with tumor cells and the advantages and disadvantages of 2D and 3D culture systems are considered.
Collapse
Affiliation(s)
- Tatyana A. Grigoreva
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia (V.G.T.)
| | | | | | | | | |
Collapse
|
9
|
Sekeroglu ZA, Sekeroglu V. A Review on Patient-derived 3D Micro Cancer Approach for Drug Screen in Personalized Cancer Medicine. Curr Cancer Drug Targets 2025; 25:118-130. [PMID: 38445692 DOI: 10.2174/0115680096285910240206044830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 03/07/2024]
Abstract
Precision medicine in oncology aims to identify an individualized treatment plan based on genomic alterations in a patient's tumor. It helps to select the most beneficial therapy for an individual patient. As it is now known that no patient's cancer is the same, and therefore, different patients may respond differently to conventional treatments, precision medicine, which replaces the one-size-fits-all approach, supports the development of tailored treatments for specific cancers of different patients. Patient-specific organoid or spheroid models as 3D cell culture models are very promising for predicting resistance to anti-cancer drugs and for identifying the most effective cancer therapy for high-throughput drug screening combined with genomic analysis in personalized medicine. Because tumor spheroids incorporate many features of solid tumors and reflect resistance to drugs and radiation, as in human cancers, they are widely used in drug screening studies. Testing patient-derived 3D cancer spheroids with some anticancer drugs based on information from molecular profiling can reveal the sensitivity of tumor cells to drugs and provide the right compounds to be effective against resistant cells. Given that many patients do not respond to standard treatments, patient-specific treatments will be more effective, less toxic. They will affect survival better compared to the standard approach used for all patients.
Collapse
Affiliation(s)
- Zulal Atlı Sekeroglu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| | - Vedat Sekeroglu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| |
Collapse
|
10
|
Patel T, Jain N. Multicellular tumor spheroids: A convenient in vitro model for translational cancer research. Life Sci 2024; 358:123184. [PMID: 39490437 DOI: 10.1016/j.lfs.2024.123184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/11/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
In the attempts to mitigate uncertainties in the results of monolayer culture for the identification of cancer therapeutic targets and compounds, there has been a growing interest in using 3D cancer spheroid models, which include tumorospheres (TSs), tissue-derived tumor spheres (TDTSs), organotypic multicellular tumor spheroids (OMSs), and multicellular tumor spheroids (MCTSs). The MCTSs, either Mono-MCTSs or Hetero-MCTSs, with or without scaffold, in particular, offer numerous advantages over other spheroid models, including easy cultivation, high reproducibility, accessibility, high throughput, controllable size, well-rounded shape, simplicity of genetic manipulation, economical and availability of various biological methods for their development. In this review, we have attempted to discuss the role of MCTSs concerning various aspects of translational cancer research, such as drug response and penetration, cell-cell interaction, and invasion and metastasis. However, the Mono-MCTSs, either scaffold-free or scaffold-based, may not adequately represent the cellular heterogeneity and complexity of clinical tumors, limiting their utility in translational cancer research. Conversely, Hetero-MCTS models, both scaffold-free and scaffold-based, show better suitability due to the presence of a similar in vivo type tumor microenvironment. Nonetheless, scaffold-based Hetero-MCTS models show batch variability and challenges in performing quantitative assays due to difficulties extracting spheroids and cells from scaffolds. Further, incorporating stromal cells with cancer cells in a more precise ratio to develop Hetero-MCTSs can enhance the model's relevance, yielding more clinically reliable outcomes for drug candidates and improving insights into tumor biology.
Collapse
Affiliation(s)
- Tushar Patel
- P D Patel Institute of Applied Sciences, Charotar University of Science and Technology (CHARUSAT), Changa 388 421, India
| | - Neeraj Jain
- Dr. K C Patel Research and Development Centre, University Research Centre(s), Charotar University of Science and Technology (CHARUSAT), Changa 388 421, India.
| |
Collapse
|
11
|
Deng J, Qin JH, Li X, Tao D, Feng Y. Establishment and drug resistance characterization of paired organoids using human primary colorectal cancer and matched tumor deposit specimens. Hum Cell 2024; 38:13. [PMID: 39495391 PMCID: PMC11534897 DOI: 10.1007/s13577-024-01139-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/13/2024] [Indexed: 11/05/2024]
Abstract
Tumor deposits (TDs) represent a specific form tumor metastasis observed in colorectal cancer (CRC). The lack of successfully established cell lines for TDs, as well as the molecular mechanisms by which TDs occur remain largely unknown. Here, we established paired CRC organoids, including a human primary cancer organoid and its TD organoid, from a 46-year-old male patient with CRC. Further analysis revealed that, compared with primary tumor-derived cells, TD-derived cells exhibited enhanced proliferative, invasive and metastatic capabilities, and increased expression of stemness-related proteins. Furthermore, the present findings also demonstrated that TD-derived cells were more resistant to oxaliplatin or 5-FU. Transcriptomic profiling and qPCR revealed that TD-derived cells exhibited more alterations in fatty acid metabolism signaling and enhanced lipid synthesis ability compared to primary tumor-derived cells. Inhibition of lipid synthesis markedly decreased resistance to oxaliplatin in TD-derived cells. Taken together, the paired organoids established using CRC primary tumor and its TD specimens will provide valuable tools to study tumorigenicity, metastasis and chemoresistance in CRC. Notably, these models will provide novel insights to study tumor heterogeneity and lipid metabolism in CRC.
Collapse
Affiliation(s)
- Jiao Deng
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Jerry H Qin
- Wuhan Britain-China Senior High School, Wuhan, 430030, China
| | - Xiaolan Li
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Deding Tao
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Yongdong Feng
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
- Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| |
Collapse
|
12
|
Lin Y, Coppo R, Onuma K, Endo H, Kondo J, Iwabuchi S, Hashimoto S, Itatani Y, Obama K, Inoue M. Growth pattern of de novo small clusters of colorectal cancer is regulated by Notch signaling at detachment. Cancer Sci 2024; 115:3648-3659. [PMID: 39300760 PMCID: PMC11531966 DOI: 10.1111/cas.16299] [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: 02/14/2024] [Revised: 07/08/2024] [Accepted: 07/17/2024] [Indexed: 09/22/2024] Open
Abstract
Cancer cell clusters have a higher capacity for metastasis than single cells, suggesting cancer cell clusters have biological properties different from those of single cells. The nature of de novo cancer cell clusters that are newly formed from tumor masses is largely unknown. Herein, we generated small cell clusters from colorectal cancer organoids and tracked the growth patterns of the clusters up to four cells. Growth patterns were classified into actively growing and poorly growing spheroids (PG). Notch signaling was robustly activated in small clusters immediately after dissociation, and Notch signaling inhibition markedly increased the proportion of PG spheroids. Only a limited number of PG spheroids grew under growth-permissive conditions in vitro, but xenograft tumors derived from Notch inhibited clusters showed growth rates comparable to those of untreated spheroids. Thus, de novo clusters are composed of cells with interchangeable growth fates, which are regulated in a context-dependent manner by Notch signaling.
Collapse
Affiliation(s)
- Yi‐Kai Lin
- Department of Clinical Bio‐resource Research and DevelopmentGraduate School of Medicine, Kyoto UniversityKyotoJapan
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Roberto Coppo
- Department of Clinical Bio‐resource Research and DevelopmentGraduate School of Medicine, Kyoto UniversityKyotoJapan
| | - Kunishige Onuma
- Department of Clinical Bio‐resource Research and DevelopmentGraduate School of Medicine, Kyoto UniversityKyotoJapan
| | - Hiroko Endo
- Department of BiochemistryOsaka International Cancer InstituteOsakaJapan
- Present address:
Carna Biosciences Inc.HyogoJapan
| | - Jumpei Kondo
- Department of Clinical Bio‐resource Research and DevelopmentGraduate School of Medicine, Kyoto UniversityKyotoJapan
- Present address:
Division of Health Sciences, Department of Molecular Biology and Clinical InvestigationGraduate School of Medicine, Osaka UniversityOsakaJapan
| | - Sadahiro Iwabuchi
- Department of Molecular PathophysiologyInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
| | - Shinichi Hashimoto
- Department of Molecular PathophysiologyInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
| | - Yoshiro Itatani
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Masahiro Inoue
- Department of Clinical Bio‐resource Research and DevelopmentGraduate School of Medicine, Kyoto UniversityKyotoJapan
| |
Collapse
|
13
|
Randall-Demllo S, Al-Qadami G, Raposo AE, Ma C, Priebe IK, Hor M, Singh R, Fung KYC. Ex Vivo Intestinal Organoid Models: Current State-of-the-Art and Challenges in Disease Modelling and Therapeutic Testing for Colorectal Cancer. Cancers (Basel) 2024; 16:3664. [PMID: 39518102 PMCID: PMC11544769 DOI: 10.3390/cancers16213664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Despite improvements in participation in population-based screening programme, colorectal cancer remains a major cause of cancer-related mortality worldwide. Targeted interventions are desirable to reduce the health and economic burden of this disease. Two-dimensional monolayers of colorectal cancer cell lines represent the traditional in vitro models for disease and are often used for diverse purposes, including the delineation of molecular pathways associated with disease aetiology or the gauging of drug efficacy. The lack of complexity in such models, chiefly the limited epithelial cell diversity and differentiation, attenuated mucus production, lack of microbial interactions and mechanical stresses, has driven interest in the development of more holistic and physiologically relevant in vitro model systems. In particular, established ex vivo patient-derived explant and patient-derived tumour xenograft models have been supplemented by progress in organoid and microfluidic organ-on-a-chip cultures. Here, we discuss the applicability of advanced culturing technologies, such as organoid systems, as models for colorectal cancer and for testing chemotherapeutic drug sensitivity and efficacy. We highlight current challenges associated with organoid technologies and discuss their future for more accurate disease modelling and personalized medicine.
Collapse
Affiliation(s)
- Sarron Randall-Demllo
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Adelaide 5000, Australia; (S.R.-D.); (G.A.-Q.)
| | - Ghanyah Al-Qadami
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Adelaide 5000, Australia; (S.R.-D.); (G.A.-Q.)
| | - Anita E. Raposo
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Westmead 2145, Australia; (A.E.R.); (C.M.)
| | - Chenkai Ma
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Westmead 2145, Australia; (A.E.R.); (C.M.)
| | - Ilka K. Priebe
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Adelaide 5000, Australia; (S.R.-D.); (G.A.-Q.)
| | - Maryam Hor
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Adelaide 5000, Australia; (S.R.-D.); (G.A.-Q.)
| | - Rajvinder Singh
- Division of Gastroenterology, Lyell McEwin Hospital, Adelaide 5112, Australia
| | - Kim Y. C. Fung
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Westmead 2145, Australia; (A.E.R.); (C.M.)
| |
Collapse
|
14
|
Airola C, Pallozzi M, Cesari E, Cerrito L, Stella L, Sette C, Giuliante F, Gasbarrini A, Ponziani FR. Hepatocellular-Carcinoma-Derived Organoids: Innovation in Cancer Research. Cells 2024; 13:1726. [PMID: 39451244 PMCID: PMC11505656 DOI: 10.3390/cells13201726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Hepatocellular carcinomas (HCCs) are highly heterogeneous malignancies. They are characterized by a peculiar tumor microenvironment and dense vascularization. The importance of signaling between immune cells, endothelial cells, and tumor cells leads to the difficult recapitulation of a reliable in vitro HCC model using the conventional two-dimensional cell cultures. The advent of three-dimensional organoid tumor technology has revolutionized our understanding of the pathogenesis and progression of several malignancies by faithfully replicating the original cancer genomic, epigenomic, and microenvironmental landscape. Organoids more closely mimic the in vivo environment and cell interactions, replicating factors such as the spatial organization of cell surface receptors and gene expression, and will probably become an important tool in the choice of therapies and the evaluation of tumor response to treatments. This review aimed to describe the ongoing and potential applications of organoids as an in vitro model for the study of HCC development, its interaction with the host's immunity, the analysis of drug sensitivity tests, and the current limits in this field.
Collapse
Affiliation(s)
- Carlo Airola
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
| | - Maria Pallozzi
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
| | - Eleonora Cesari
- GSTeP Organoids Research Core Facility, Fondazione Policlinico A. Gemelli, 00168 Rome, Italy; (E.C.); (C.S.)
| | - Lucia Cerrito
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
| | - Leonardo Stella
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
| | - Claudio Sette
- GSTeP Organoids Research Core Facility, Fondazione Policlinico A. Gemelli, 00168 Rome, Italy; (E.C.); (C.S.)
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Felice Giuliante
- Department of Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
15
|
Esposito A, Ferraresi A, Vallino L, Garavaglia B, Dhanasekaran DN, Isidoro C. Three-Dimensional In Vitro Cell Cultures as a Feasible and Promising Alternative to Two-Dimensional and Animal Models in Cancer Research. Int J Biol Sci 2024; 20:5293-5311. [PMID: 39430243 PMCID: PMC11488579 DOI: 10.7150/ijbs.96469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/25/2024] [Indexed: 10/22/2024] Open
Abstract
Cancer represents one of the diseases with the highest mortality rate worldwide. The burden of cancer continues to increase, not only affecting the health-related quality of life of patients but also causing an elevated global financial impact. The complexity and heterogeneity of cancer pose significant challenges in research and clinical practice, contributing to increase the failure rate of clinical trials for antitumoral drugs. This is partially due to the fact that preclinical models still present important limitations in faithfully recapitulating human tumors to serve as reliable indicators of drug effectiveness. Up to now, research and development strategies employ expensive animal models (including the so-called "humanized mice") that not only raise ethical concerns, but also frequently fail to accurately predict responses to anticancer drugs because they do not faithfully replicate human physiology as well as the patient's tumor microenvironment. On the other side, traditional two-dimensional (2D) cell cultures fail to adequately reproduce the structural organization of tumor and the cellular heterogeneity found in vivo. The growing necessity to develop more accurate cancer models has increasingly emphasized the importance of three-dimensional (3D) in vitro cell cultures, such as cancer-derived spheroids and organoids, as promising alternatives to bridge the gap between 2D and animal models. In this review, we provide a brief overview focusing on 3D in vitro cell cultures as preclinical models capable of properly reproducing the tissue organization, biological composition, and complexity of in vivo tumors in a fine-tuned microenvironment. Despite their limitations, these models collectively enhance our understanding of the mechanisms underlying cancer and may offer the potential for a more reliable assessment of drug efficacy before clinical testing and, consequently, improve therapeutic outcomes for cancer patients.
Collapse
Affiliation(s)
- Andrea Esposito
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Letizia Vallino
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Beatrice Garavaglia
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Danny N. Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| |
Collapse
|
16
|
Luo X, Gong Y, Gong Z, Fan K, Suo T, Liu H, Ni X, Ni X, Abudureyimu M, Liu H. Liver and bile duct organoids and tumoroids. Biomed Pharmacother 2024; 178:117104. [PMID: 39024834 DOI: 10.1016/j.biopha.2024.117104] [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: 03/28/2024] [Revised: 06/23/2024] [Accepted: 07/07/2024] [Indexed: 07/20/2024] Open
Abstract
Organoids refer to 3D cultures established to recapitulate histology, pathology, architecture, and genetic traits of various organs and tissues in the body, thereby replacing 2D cell cultures, xenograft, and animal models. Organoids form a 3D in vitro mimic of original tissues like the liver and are derived from embryonic or adult tissue stem cells. Liver and bile duct tumor organoids, also called, tumoroids capture genetic diversity, cellular, and pathophysiological properties of original tumors. Moreover, co-culture techniques along with genetic modulation of organoids allow for using tumoroids in liver and bile duct cancer research and drug screening/testing. Therefore, tumoroids are promising platforms for studying liver and bile duct cancer, which paves the way for the new era of personalized therapies. In the current review, we aimed to discuss liver and bile duct organoids with special emphasis on tumoroids and their applications, advantages, and shortcomings.
Collapse
Affiliation(s)
- Xuanming Luo
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China; Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
| | - Yuda Gong
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China
| | - Zijun Gong
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China
| | - Kun Fan
- Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
| | - Tao Suo
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China
| | - Han Liu
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China
| | - Xiaoling Ni
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China
| | - Xiaojian Ni
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China
| | - Miyesaier Abudureyimu
- Cardiovascular Department, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China.
| | - Houbao Liu
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China; Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
17
|
Rahman MM, Wells G, Rantala JK, Helleday T, Muthana M, Danson SJ. In-vitro assays for immuno-oncology drug efficacy assessment and screening for personalized cancer therapy: scopes and challenges. Expert Rev Clin Immunol 2024; 20:821-838. [PMID: 38546609 DOI: 10.1080/1744666x.2024.2336583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Immunotherapies have revolutionized cancer treatment, but often fail to produce desirable therapeutic outcomes in all patients. Due to the inter-patient heterogeneity and complexity of the tumor microenvironment, personalized treatment approaches are gaining demand. Researchers have long been using a range of in-vitro assays including 2D models, organoid co-cultures, and cancer-on-a-chip platforms for cancer drug screening. A comparative analysis of these assays with their suitability, high-throughput capacity, and clinical translatability is required for optimal translational use. AREAS COVERED The review summarized in-vitro platforms with their comparative advantages and limitations including construction strategies, and translational potential for immuno-oncology drug efficacy assessment. We also discussed end-point analysis strategies so that researchers can contextualize their usefulness and optimally design experiments for personalized immunotherapy efficacy prediction. EXPERT OPINION Researchers developed several in-vitro platforms that can provide information on personalized immunotherapy efficacy from different angles. Image-based assays are undoubtedly more suitable to gather a wide range of information including cellular morphology and phenotypical behaviors but need significant improvement to overcome issues including background noise, sample preparation difficulty, and long duration of experiment. More studies and clinical trials are needed to resolve these issues and validate the assays before they can be used in real-life scenarios.
Collapse
Affiliation(s)
- Md Marufur Rahman
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Directorate General of Health Services, Dhaka, Bangladesh
| | - Greg Wells
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| | - Juha K Rantala
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Misvik Biology Ltd, Turku, Finland
| | - Thomas Helleday
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Department of Oncology-Pathology, Karolinska Institutet, Huddinge, Sweden
| | - Munitta Muthana
- Nanobug Oncology Sheffield, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| | - Sarah J Danson
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| |
Collapse
|
18
|
Reuvers TGA, Grandia V, Brandt RMC, Arab M, Maas SLN, Bos EM, Nonnekens J. Investigating the Radiobiological Response to Peptide Receptor Radionuclide Therapy Using Patient-Derived Meningioma Spheroids. Cancers (Basel) 2024; 16:2515. [PMID: 39061156 PMCID: PMC11275064 DOI: 10.3390/cancers16142515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) using 177Lu-DOTA-TATE has recently been evaluated for the treatment of meningioma patients. However, current knowledge of the underlying radiation biology is limited, in part due to the lack of appropriate in vitro models. Here, we demonstrate proof-of-concept of a meningioma patient-derived 3D culture model to assess the short-term response to radiation therapies such as PRRT and external beam radiotherapy (EBRT). We established short-term cultures (1 week) for 16 meningiomas with high efficiency and yield. In general, meningioma spheroids retained characteristics of the parental tumor during the initial days of culturing. For a subset of tumors, clear changes towards a more aggressive phenotype were visible over time, indicating that the culture method induced dedifferentiation of meningioma cells. To assess PRRT efficacy, we demonstrated specific uptake of 177Lu-DOTA-TATE via somatostatin receptor subtype 2 (SSTR2), which was highly overexpressed in the majority of tumor samples. PRRT induced DNA damage which was detectable for an extended timeframe as compared to EBRT. Interestingly, levels of DNA damage in spheroids after PRRT correlated with SSTR2-expression levels of parental tumors. Our patient-derived meningioma culture model can be used to assess the short-term response to PRRT and EBRT in radiobiological studies. Further improvement of this model should pave the way towards the development of a relevant culture model for assessment of the long-term response to radiation and, potentially, individual patient responses to PRRT and EBRT.
Collapse
Affiliation(s)
- Thom G A Reuvers
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Vivian Grandia
- Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Renata M C Brandt
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Majd Arab
- Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Sybren L N Maas
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Eelke M Bos
- Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Julie Nonnekens
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
19
|
Tabata S, Endo H, Makinoshima H, Soga T, Inoue M. The γ-glutamyl cycle serves as an amino acids supply system in colorectal cancer organoids under chronic hypoxia. Biochem Biophys Res Commun 2024; 714:149977. [PMID: 38663093 DOI: 10.1016/j.bbrc.2024.149977] [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: 03/19/2024] [Revised: 04/13/2024] [Accepted: 04/21/2024] [Indexed: 05/07/2024]
Abstract
Malignant tumors are characterized by a hypoxic microenvironment, and metabolic reprogramming is necessary to ensure energy production and oxidative stress resistance. Although the microenvironmental properties of tumors vary under acute and chronic hypoxia, studies on chronic hypoxia-induced metabolic changes are limited. In the present study, we performed a comprehensive metabolic analysis in a chronic hypoxia model using colorectal cancer (CRC) organoids, and identified an amino acid supply system through the γ-glutamyl cycle, a glutathione recycling pathway. We analyzed the metabolic changes caused by hypoxia over time and observed that chronic hypoxia resulted in an increase in 5-oxoproline and a decrease in oxidized glutathione (GSSG) compared to acute hypoxia. These findings suggest that chronic hypoxia induces metabolic changes in the γ-glutamyl cycle. Moreover, inhibition of the γ-glutamyl cycle via γ-glutamyl cyclotransferase (GGCT) and γ-glutamyl transferase 1 (GGT1) knockdown significantly reversed chronic hypoxia-induced upregulation of 5-oxoproline and several amino acids. Notably, GGT1 knockdown downregulated the intracellular levels of γ-glutamyl amino acids. Conclusively, these results indicate that the γ-glutamyl cycle serves as an amino acid supply system in CRC under chronic hypoxia, which provides fresh insight into cancer metabolism under chronic hypoxia.
Collapse
Affiliation(s)
- Sho Tabata
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Tsuruoka, 997-0052, Japan; Tsuruoka Metabolomics Laboratory, National Cancer Center, Tsuruoka, 997-0052, Japan; Shonai Regional Industry Promotion Center, Tsuruoka, 997-0052, Japan; Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, 277-8577, Japan.
| | - Hiroko Endo
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Hideki Makinoshima
- Tsuruoka Metabolomics Laboratory, National Cancer Center, Tsuruoka, 997-0052, Japan; Shonai Regional Industry Promotion Center, Tsuruoka, 997-0052, Japan; Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, 277-8577, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Tsuruoka, 997-0052, Japan; Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, 108-8345 Japan
| | - Masahiro Inoue
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan; Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| |
Collapse
|
20
|
Yu S, Zhang L, Yang Y, Wang M, Liu T, Ji W, Liu Y, Lv H, Zhao Y, Chen X, Hu T. Polydopamine-Based Resveratrol-Hyaluronidase Nanomedicine Inhibited Pancreatic Cancer Cell Invasive Phenotype in Hyaluronic Acid Enrichment Tumor Sphere Model. ACS Pharmacol Transl Sci 2024; 7:1013-1022. [PMID: 38633596 PMCID: PMC11020062 DOI: 10.1021/acsptsci.3c00304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/13/2023] [Accepted: 12/25/2023] [Indexed: 04/19/2024]
Abstract
The dense storm microenvironment formed by an excessively cross-linked extracellular matrix, such as hyaluronic acid and collagens, serves as a major barrier that prevents drugs from reaching the deeper tumor. Current traditional two-dimensional (2D) cultures are not capable of modeling this drug delivery barrier in vitro. Thus, tumor spheroids have become increasingly important in cancer research due to their three-dimensional structure. Currently, various methods have been developed to construct tumor spheroids. However, there are still challenges, such as lengthy construction time, complex composition of added growth factors, and high cultivation costs. To address this technical bottleneck, our study combined the GelMA hydrogel system to develop a rapid and high-yield method for tumor spheroids generation. Additionally, we proposed an evaluation scheme to assess the effects of drugs on tumor spheroids. Building on the hyaluronic acid-rich pathological tumor microenvironment, we constructed a resveratrol-loaded nano-drug delivery system with tumor stroma modulation capability and used a three-dimensional (3D) tumor sphere model to simulate in vivo tumor conditions. This process was utilized to completely evaluate the ability of the nano-drug delivery system to enhance the deep penetration of resveratrol in the tumor microenvironment, providing new insights into future oncology drug screening, efficacy assessment, and drug delivery methods.
Collapse
Affiliation(s)
- Shuo Yu
- Department
of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department
of General Surgery, The Second Affiliated
Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
- Bioinspired
Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an, Shaanxi 710000, China
| | - Lu Zhang
- National
& Local Joint Engineering Research Center of Biodiagnosis and
Biotherapy, The Second Affiliated Hospital
of Xi’an Jiaotong University, Xi’an 710004, China
| | - Yanshen Yang
- Bioinspired
Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an, Shaanxi 710000, China
| | - Meijuan Wang
- Department
of Anesthesia, Guangdong Provincial People’s
Hospital, Guangzhou 510080, China
| | - Tingting Liu
- Department
of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Wenwen Ji
- Department
of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Yang Liu
- Department
of General Surgery, The Second Affiliated
Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Hao Lv
- Department
of General Surgery, The Second Affiliated
Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Yang Zhao
- National
& Local Joint Engineering Research Center of Biodiagnosis and
Biotherapy, The Second Affiliated Hospital
of Xi’an Jiaotong University, Xi’an 710004, China
| | - Xi Chen
- National
& Local Joint Engineering Research Center of Biodiagnosis and
Biotherapy, The Second Affiliated Hospital
of Xi’an Jiaotong University, Xi’an 710004, China
| | - Tinghua Hu
- Department
of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| |
Collapse
|
21
|
Radu P, Zurzu M, Tigora A, Paic V, Bratucu M, Garofil D, Surlin V, Munteanu AC, Coman IS, Popa F, Strambu V, Ramboiu S. The Impact of Cancer Stem Cells in Colorectal Cancer. Int J Mol Sci 2024; 25:4140. [PMID: 38673727 PMCID: PMC11050141 DOI: 10.3390/ijms25084140] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Despite incessant research, colorectal cancer (CRC) is still one of the most common causes of fatality in both men and women worldwide. Over time, advancements in medical treatments have notably enhanced the survival rates of patients with colorectal cancer. Managing metastatic CRC involves a complex tradeoff between the potential benefits and adverse effects of treatment, considering factors like disease progression, treatment toxicity, drug resistance, and the overall impact on the patient's quality of life. An increasing body of evidence highlights the significance of the cancer stem cell (CSC) concept, proposing that CSCs occupy a central role in triggering cancer. CSCs have been a focal point of extensive research in a variety of cancer types, including CRC. Colorectal cancer stem cells (CCSCs) play a crucial role in tumor initiation, metastasis, and therapy resistance, making them potential treatment targets. Various methods exist for isolating CCSCs, and understanding the mechanisms of drug resistance associated with them is crucial. This paper offers an overview of the current body of research pertaining to the comprehension of CSCs in colorectal cancer.
Collapse
Affiliation(s)
- Petru Radu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Mihai Zurzu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Anca Tigora
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Vlad Paic
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Mircea Bratucu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Dragos Garofil
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Valeriu Surlin
- Sixth Department of Surgery, University of Medicine and Pharmacy of Craiova, Craiova Emergency Clinical 7 Hospital, 200642 Craiova, Romania; (V.S.); (A.C.M.); (S.R.)
| | - Alexandru Claudiu Munteanu
- Sixth Department of Surgery, University of Medicine and Pharmacy of Craiova, Craiova Emergency Clinical 7 Hospital, 200642 Craiova, Romania; (V.S.); (A.C.M.); (S.R.)
| | - Ionut Simion Coman
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
- General Surgery Department, “Bagdasar-Arseni” Clinical Emergency Hospital, 12 Berceni Road, 041915 Bucharest, Romania
| | - Florian Popa
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Victor Strambu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Sandu Ramboiu
- Sixth Department of Surgery, University of Medicine and Pharmacy of Craiova, Craiova Emergency Clinical 7 Hospital, 200642 Craiova, Romania; (V.S.); (A.C.M.); (S.R.)
| |
Collapse
|
22
|
Sasagawa S, Kumai J, Wakamatsu T, Yui Y. Improvement of histone deacetylase inhibitor efficacy by SN38 through TWIST1 suppression in synovial sarcoma. CANCER INNOVATION 2024; 3:e113. [PMID: 38946933 PMCID: PMC11212284 DOI: 10.1002/cai2.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/14/2023] [Accepted: 12/22/2023] [Indexed: 07/02/2024]
Abstract
Background Synovial sarcoma (SS) is an SS18-SSX fusion gene-driven soft tissue sarcoma with mesenchymal characteristics, associated with a poor prognosis due to frequent metastasis to a distant organ, such as the lung. Histone deacetylase (HDAC) inhibitors (HDACis) are arising as potent molecular targeted drugs, as HDACi treatment disrupts the SS oncoprotein complex, which includes HDACs, in addition to general HDACi effects. To provide further molecular evidence for the advantages of HDACi treatment and its limitations due to drug resistance induced by the microenvironment in SS cells, we examined cellular responses to HDACi treatment in combination with two-dimensional (2D) and 3D culture conditions. Methods Using several SS cell lines, biochemical and cell biological assays were performed with romidepsin, an HDAC1/2 selective inhibitor. SN38 was concomitantly used as an ameliorant drug with romidepsin treatment. Cytostasis, apoptosis induction, and MHC class I polypeptide-related sequence A/B (MICA/B) induction were monitored to evaluate the drug efficacy. In addition to the conventional 2D culture condition, spheroid culture was adopted to evaluate the influence of cell-mass microenvironment on chemoresistance. Results By monitoring the cellular behavior with romidepsin and/or SN38 in SS cells, we observed that responsiveness is diverse in each cell line. In the apoptotic inducible cells, co-treatment with SN38 enhanced cell death. In nonapoptotic inducible cells, cytostasis and MICA/B induction were observed, and SN38 improved MICA/B induction further. As a novel efficacy of SN38, we revealed TWIST1 suppression in SS cells. In the spheroid (3D) condition, romidepsin efficacy was severely restricted in TWIST1-positive cells. We demonstrated that TWIST1 downregulation restored romidepsin efficacy even in spheroid form, and concomitant SN38 treatment along with romidepsin reproduced the reaction. Conclusions The current study demonstrated the benefits and concerns of using HDACi for SS treatment in 2D and 3D culture conditions and provided molecular evidence that concomitant treatment with SN38 can overcome drug resistance to HDACi by suppressing TWIST1 expression.
Collapse
Affiliation(s)
- Satoru Sasagawa
- Molecular Biology Laboratory, Research InstituteNozaki Tokushukai HospitalDaitoOsakaJapan
| | - Jun Kumai
- Sarcoma Treatment Laboratory, Research InstituteNozaki Tokushukai HospitalDaitoOsakaJapan
| | - Toru Wakamatsu
- Department of Musculoskeletal Oncology ServiceOsaka International Cancer InstituteOsakaJapan
| | - Yoshihiro Yui
- Sarcoma Treatment Laboratory, Research InstituteNozaki Tokushukai HospitalDaitoOsakaJapan
| |
Collapse
|
23
|
Masuda M, Iida K, Iwabuchi S, Tanaka M, Kubota S, Uematsu H, Onuma K, Kukita Y, Kato K, Kamiura S, Nakajima A, Coppo R, Kanda M, Yoshino K, Ueda Y, Morii E, Kimura T, Kondo J, Okada-Hatakeyama M, Hashimoto S, Inoue M. Clonal Origin and Lineage Ambiguity in Mixed Neuroendocrine Carcinoma of the Uterine Cervix. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:415-429. [PMID: 38103888 DOI: 10.1016/j.ajpath.2023.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/23/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023]
Abstract
Small-cell neuroendocrine carcinoma (SCNEC) of the cervix is a rare disease characterized by a high incidence of mixed tumors with other types of cancer. The mechanism underlying this mixed phenotype is not well understood. This study established a panel of organoid lines from patients with SCNEC of the cervix and ultimately focused on one line, which retained a mixed tumor phenotype, both in vitro and in vivo. Histologically, both organoids and xenograft tumors showed distinct differentiation into either SCNEC or adenocarcinoma in some regions and ambiguous differentiation in others. Tracking single cells indicated the existence of cells with bipotential differentiation toward SCNEC and adenocarcinomas. Single-cell transcriptional analysis identified three distinct clusters: SCNEC-like, adenocarcinoma-like, and a cluster lacking specific differentiation markers. The expression of neuroendocrine markers was enriched in the SCNEC-like cluster but not exclusively. Human papillomavirus 18 E6 was enriched in the SCNEC-like cluster, which showed higher proliferation and lower levels of the p53 pathway. After treatment with anticancer drugs, the expression of adenocarcinoma markers increased, whereas that of SCNEC decreased. Using a reporter system for keratin 19 expression, changes in the differentiation of each cell were shown to be associated with the shift in differentiation induced by drug treatment. These data suggest that mixed SCNEC/cervical tumors have a clonal origin and are characterized by an ambiguous and flexible differentiation state.
Collapse
Affiliation(s)
- Masamune Masuda
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Keita Iida
- Laboratory of Cell Systems, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Sadahiro Iwabuchi
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Mie Tanaka
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Satoshi Kubota
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan; Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Hiroyuki Uematsu
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kunishige Onuma
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoji Kukita
- Department of Molecular and Medical Genetics, Osaka International Cancer Institute, Osaka, Japan
| | - Kikuya Kato
- Department of Molecular and Medical Genetics, Osaka International Cancer Institute, Osaka, Japan
| | - Shoji Kamiura
- Department of Gynecology, Osaka International Cancer Institute, Osaka, Japan
| | - Aya Nakajima
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan; Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Roberto Coppo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mizuki Kanda
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kiyoshi Yoshino
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yutaka Ueda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jumpei Kondo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | | | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan.
| |
Collapse
|
24
|
Rodrigues DB, Reis RL, Pirraco RP. Modelling the complex nature of the tumor microenvironment: 3D tumor spheroids as an evolving tool. J Biomed Sci 2024; 31:13. [PMID: 38254117 PMCID: PMC10804490 DOI: 10.1186/s12929-024-00997-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer remains a serious burden in society and while the pace in the development of novel and more effective therapeutics is increasing, testing platforms that faithfully mimic the tumor microenvironment are lacking. With a clear shift from animal models to more complex in vitro 3D systems, spheroids emerge as strong options in this regard. Years of development have allowed spheroid-based models to better reproduce the biomechanical cues that are observed in the tumor-associated extracellular matrix (ECM) and cellular interactions that occur in both a cell-cell and cell-ECM manner. Here, we summarize some of the key cellular interactions that drive tumor development, progression and invasion, and how successfully are these interactions recapitulated in 3D spheroid models currently in use in the field. We finish by speculating on future advancements in the field and on how these can shape the relevance of spherical 3D models for tumor modelling.
Collapse
Affiliation(s)
- Daniel B Rodrigues
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, 4805-017, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, 4805-017, Guimarães, Portugal
| | - Rogério P Pirraco
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal.
- ICVS/3B's, PT Government Associate Laboratory, Braga, 4805-017, Guimarães, Portugal.
| |
Collapse
|
25
|
Zhang D, Tang D, Liu PT, Tao L, Lu LM. Isolation of tumor stem-like cells from primary laryngeal squamous cell carcinoma cells (FD-LS-6). Hum Cell 2024; 37:323-336. [PMID: 37759147 DOI: 10.1007/s13577-023-00984-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023]
Abstract
The development of efficient treatments for laryngeal squamous cell carcinoma (LSCC) is hindered by the lack of applicable tumor cell lines and animal models of the disease, especially those related to cancer stem-like cells (CSCs). CSCs play critical roles in tumor propagation and pathogenesis whereas no CSCs lines have been developed to date. In this study, we establish an LSCC cell line (FD-LS-6) from primary LSCC tumor tissue (not experienced single-cell cloning) and adapted a culturing condition for the expansion of potential stem cells (EPSCs) to isolate CSCs from FD-LS-6. We successfully derived novel CSCs and named them as LSCC sphere-forming cells (LSCSCs) which were subsequently characterized for their CSC properties. We showed that LSCSCs shared many properties of CSCs, including CSC marker, robust self-renewal capacity, tumorigenesis ability, potential to generate other cell types such as adipocytes and osteoblasts, and resistance to chemotherapy. Compared to parental cells, LSCSCs were significantly more potent in forming tumors in vivo in mice and more resistant to chemotherapy. LSCSCs have higher expressions of epithelial-mesenchymal transition proteins and chemotherapy resistance factors, and exhibit an activated COX2/PEG2 signaling pathway. Altogether, our work establishes the first CSCs of LSCC (FD-LS-6) and provides a tool to study tumorigenesis and metastasis of LSCC and help the development of anticancer therapies.
Collapse
Affiliation(s)
- Duo Zhang
- Department of Otolaryngology-HNS, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University School of Medicine, 83 Fenyang Road, Shanghai, 200031, China
- Department of Pudong Hospital, Fudan University School of Medicine, 2800 Gongwei Road, Shanghai, 201300, China
| | - Di Tang
- Department of Otolaryngology-HNS, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University School of Medicine, 83 Fenyang Road, Shanghai, 200031, China
- Department of Pudong Hospital, Fudan University School of Medicine, 2800 Gongwei Road, Shanghai, 201300, China
| | - Pen-Tao Liu
- School of Biomedical Sciences, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 5 Sassoon Road, Hong Kong, China
- Centre for Translational Stem Cell Biology, Science and Technology Park, 6-8 Harbour Road, Hong Kong, China
| | - Lei Tao
- Department of Otolaryngology-HNS, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University School of Medicine, 83 Fenyang Road, Shanghai, 200031, China.
- Department of Pudong Hospital, Fudan University School of Medicine, 2800 Gongwei Road, Shanghai, 201300, China.
| | - Li-Ming Lu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| |
Collapse
|
26
|
Muscatello LV, Frabetti S, Avallone G, Gobbo F, Pasquini A, D'Annunzio G, Pisoni L, Marconato L, Terragni R, De Biase D, Candini O, Sarli G. Morphologic, phenotypic, and genotypic similarities between primary tumors and corresponding 3D cell cultures grown in a repeatable system-preliminary results. BMC Vet Res 2023; 19:263. [PMID: 38071286 PMCID: PMC10709889 DOI: 10.1186/s12917-023-03834-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Three-dimensional (3D) cell cultures are the new frontier for reproducing the tumor micro-environment in vitro. The aims of the study were (1) to establish primary 3D cell cultures from canine spontaneous neoplasms and (2) to demonstrate the morphological, phenotypic and genotypic similarities between the primary canine neoplasms and the corresponding 3D cultures, through the expression of tumor differentiation markers. RESULTS Seven primary tumors were collected, including 4 carcinomas and 3 soft tissue sarcomas. 3D cell cultures reproduced the morphological features of the primary tumors and showed an overlapping immunophenotype of the primary epithelial tumors. Immunohistochemistry demonstrated the growth of stromal cells and macrophages admixed with the neoplastic epithelial component, reproducing the tumor microenvironment. Mesenchymal 3D cultures reproduced the immunophenotype of the primary tumor completely in 2 out of 3 examined cases while a discordant expression was documented for a single marker in one case. No single nucleotide variants or small indel were detected in TP53 or MDM2 genes, both in primary tumors and in 3D cell cultures specimens. In one sample, MDM2 amplicons were preferentially increased in number compared to TP53 ones, indicating amplification of MDM2, detectable both in the primary tumor and in the corresponding cell culture specimen. CONCLUSION Here we demonstrate a good cell morphology, phenotype and genetic profile overlap between primary tumors and the corresponding 3D cultures grown in a repeatable system.
Collapse
Affiliation(s)
- Luisa Vera Muscatello
- Department of Veterinary Medical Sciences, University of Bologna, I-40064, Ozzano dell'Emilia, BO, Italy.
| | | | - Giancarlo Avallone
- Department of Veterinary Medical Sciences, University of Bologna, I-40064, Ozzano dell'Emilia, BO, Italy
| | - Francesca Gobbo
- Department of Veterinary Medical Sciences, University of Bologna, I-40064, Ozzano dell'Emilia, BO, Italy
| | - Arianna Pasquini
- Department of Veterinary Medical Sciences, University of Bologna, I-40064, Ozzano dell'Emilia, BO, Italy
| | - Giulia D'Annunzio
- Department of Veterinary Medical Sciences, University of Bologna, I-40064, Ozzano dell'Emilia, BO, Italy
- Experimental Zooprophylactic Institute of Lombardia and Emilia Romagna, I-25124, Brescia, Italy
| | - Luciano Pisoni
- Department of Veterinary Medical Sciences, University of Bologna, I-40064, Ozzano dell'Emilia, BO, Italy
| | - Laura Marconato
- Department of Veterinary Medical Sciences, University of Bologna, I-40064, Ozzano dell'Emilia, BO, Italy
| | | | - Dario De Biase
- Department of Pharmacy and BiotechnologyUniversity of Bologna, I-40127, Bologna, Italy
| | | | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, I-40064, Ozzano dell'Emilia, BO, Italy
| |
Collapse
|
27
|
Uematsu H, Saito C, Kondo J, Onuma K, Coppo R, Mori Y, Muto M, Kikawa Y, Tada M, Sugie T, Inoue M. De-differentiation in cultures of organoids from luminal-type breast cancer is restored by inhibition of NOTCH signaling. Hum Cell 2023; 36:2099-2112. [PMID: 37634223 DOI: 10.1007/s13577-023-00975-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/20/2023] [Indexed: 08/29/2023]
Abstract
Estrogen receptor (ER) expression in breast cancer can change during progression and the treatment, but the mechanism has not been well studied. In this study, we successfully prepared organoids from samples obtained from 33 luminal-type breast cancer patients and studied their ER expression. The expression status was well maintained in primary organoids, whereas it decreased after passaging in most of the cases. In fact, the studied organoid lines were classified into those that retained a high level of ER expression (9%), those that completely lost it (9%), and those that repressed it to varying degrees (82%). In some cases, the ER expression was suddenly and drastically decreased after passaging. Marker protein immunohistochemistry revealed that after passaging, the differentiation status shifted from a luminal- to a basal-like status. Differentially expressed genes suggested the activation of NOTCH signaling in the passaged organoids, wherein a NOTCH inhibitor was able to substantially rescue the decreased ER expression and alter the differentiation status. Our findings suggest that the differentiation status of luminal-type cancer cells is quite flexible, and that by inhibiting the NOTCH signaling we can preserve the differentiation status of luminal-type breast cancer organoids.
Collapse
Affiliation(s)
- Hiroyuki Uematsu
- Department of Clinical Bio-Resource Research and Development, Graduate School of Medicine, Kyoto University, 46-29, Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8304, Japan
- KBBM Inc, 46-29, Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8304, Japan
| | - Chieko Saito
- Department of Clinical Bio-Resource Research and Development, Graduate School of Medicine, Kyoto University, 46-29, Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8304, Japan
- KBBM Inc, 46-29, Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8304, Japan
| | - Jumpei Kondo
- Department of Clinical Bio-Resource Research and Development, Graduate School of Medicine, Kyoto University, 46-29, Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8304, Japan
- Division of Health Sciences, Department of Molecular Biology and Clinical Investigation, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kunishige Onuma
- Department of Clinical Bio-Resource Research and Development, Graduate School of Medicine, Kyoto University, 46-29, Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8304, Japan
| | - Roberto Coppo
- Department of Clinical Bio-Resource Research and Development, Graduate School of Medicine, Kyoto University, 46-29, Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8304, Japan
| | - Yukiko Mori
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yuichiro Kikawa
- Department of Breast Surgery, Kansai Medical University, Hirakata, Osaka, 573-1191, Japan
| | - Manami Tada
- Department of Breast Surgery, Kansai Medical University, Hirakata, Osaka, 573-1191, Japan
| | - Tomoharu Sugie
- Department of Breast Surgery, Kansai Medical University, Hirakata, Osaka, 573-1191, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-Resource Research and Development, Graduate School of Medicine, Kyoto University, 46-29, Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8304, Japan.
- Department of Clinical Bio-Resource Research and Development, Graduate School of Medicine, Kyoto University, Med-Pharm Collaboration Building 503, Shimoadachi-cho 46-29, Sakyou-ku, Kyoto, 606-8304, Japan.
| |
Collapse
|
28
|
Zhang Y, Huo J, Yu S, Feng W, Tuersun A, Chen F, Lv Z, Liu W, Zhao J, Xu Z, Lu A, Zong Y. Colorectal cancer tissue-originated spheroids reveal tumor intrinsic signaling pathways and mimic patient clinical chemotherapeutic response as a rapid and valid model. Biomed Pharmacother 2023; 167:115585. [PMID: 37774672 DOI: 10.1016/j.biopha.2023.115585] [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: 07/19/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023] Open
Abstract
Locally advanced colorectal cancer requires preoperative chemotherapy to reduce local recurrence and metastasis rates, but it remains difficult to predict the tumor will be sensitive to which treatments. The patient-derived organoids (PDOs) are considered an effective platform for predicting tumor drug responses in precision oncology. However, it has the limitation of being time-consuming in practical applications, especially in neoadjuvant treatment. Here we used cancer tissue-originated spheroids (CTOS) method to establish organoids from a heterogeneous population of colorectal cancer specimens, and evaluated the capacity of CTOS to predict clinical drug responses. By analyzing the relationship of the activities of drug-treated CTOS, drug targets and target-related pathways, tumor intrinsic effective-target-related pathways can be identified. These pathways were highly matched to the abnormal pathways indicated by whole-exome sequencing. Based on this, we used half effective concentration gradients to classify CTOS as sensitive or resistant to chemotherapy regimens within a week, for predicting neoadjuvant treatment outcomes for colorectal cancer patients. The drug sensitivity test results are highly matched to the clinical responses to treatment in individual patients. Thus, our data suggested that CTOS models can be effectively screened ex vivo to identify pathways sensitive to chemotherapies. These data also supported organoid research for personalized clinical medication guidance immediately after diagnosis in patients with advanced colorectal cancer.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianting Huo
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Suyue Yu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenqing Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Abudumaimaitijiang Tuersun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fangqian Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zeping Lv
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wangyi Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingkun Zhao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhuoqing Xu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Aiguo Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Yaping Zong
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
29
|
Freeburg NF, Peterson N, Ruiz DA, Gladstein AC, Feldser DM. Metastatic Competency and Tumor Spheroid Formation Are Independent Cell States Governed by RB in Lung Adenocarcinoma. CANCER RESEARCH COMMUNICATIONS 2023; 3:1992-2002. [PMID: 37728504 PMCID: PMC10545537 DOI: 10.1158/2767-9764.crc-23-0172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/08/2023] [Accepted: 09/06/2023] [Indexed: 09/21/2023]
Abstract
Inactivation of the retinoblastoma (RB) tumor suppressor in lung adenocarcinoma is associated with the rapid acquisition of metastatic ability and the loss of lung cell lineage commitment. We previously showed that restoration of RB in advanced lung adenocarcinomas in the mouse was correlated with a decreased frequency of lineage decommitted tumors and overt metastases. To identify a causal relationship for RB and its role in reprogramming lineage commitment and reducing metastatic competency in lung adenocarcinoma, we developed multiple tumor spheroid forming lines where RB restoration could be achieved after characterization of the degree of each spheroid's lineage commitment and metastatic ability. Surprisingly, we discovered that RB inactivation dramatically promoted tumor spheroid forming potential in tumors that arise in the KrasLSL-G12D/+; p53flox/flox lung adenocarcinoma model. However, RB reactivation had no effect on the maintenance of tumor spheroid lines once established. In addition, we show that RB-deficient tumor spheroid lines are not uniformly metastatically competent but are equally likely to be nonmetastatic. Interestingly, unlike tumor spheroid maintenance, RB restoration could functionally revert metastatic tumor spheroids to a nonmetastatic cell state. Thus, strategies to reinstate RB pathway activity in lung cancer may reverse metastatic ability and have therapeutic potential. Finally, the acquisition of tumor spheroid forming potential reflects underlying cell state plasticity, which is often predictive of, or even conflated with metastatic ability. Our data support that each is a discrete cell state restricted by RB and question the suitability of tumor spheroid models for their predictive potential of advanced metastatic tumor cell states. SIGNIFICANCE Members of the RB pathway are frequently mutated in lung adenocarcinoma. We show that RB regulates cell state plasticity, tumor spheroid formation, and metastatic competency. Our data indicate that these are independent states where spheroid formation is distinct from metastatic competency. Thus, we caution against conflating spheroid formation and other signs of cell state plasticity with advanced metastatic cell states. Nevertheless, our work supports clinical strategies to reactivate RB pathways.
Collapse
Affiliation(s)
- Nelson F. Freeburg
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, Pennsylvania
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nia Peterson
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Dain A. Ruiz
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amy C. Gladstein
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, Pennsylvania
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David M. Feldser
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, Pennsylvania
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
30
|
Suezawa T, Sasaki N, Yukawa Y, Assan N, Uetake Y, Onuma K, Kamada R, Tomioka D, Sakurai H, Katayama R, Inoue M, Matsusaki M. Ultra-Rapid and Specific Gelation of Collagen Molecules for Transparent and Tough Gels by Transition Metal Complexation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302637. [PMID: 37697642 PMCID: PMC10602541 DOI: 10.1002/advs.202302637] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/05/2023] [Indexed: 09/13/2023]
Abstract
Collagen is the most abundant protein in the human body and one of the main components of stromal tissues in tumors which have a high elastic modulus of over 50 kPa. Although collagen has been widely used as a cell culture scaffold for cancer cells, there have been limitations when attempting to fabricate a tough collagen gel with cells like a cancer stroma. Here, rapid gelation of a collagen solution within a few minutes by transition metal complexation is demonstrated. Type I collagen solution at neutral pH shows rapid gelation with a transparency of 81% and a high modulus of 1,781 kPa by mixing with K2 PtCl4 solution within 3 min. Other transition metal ions also show the same rapid gelation, but not basic metal ions. Interestingly, although type I to IV collagen molecules show rapid gelation, other extracellular matrices do not exhibit this phenomenon. Live imaging of colon cancer organoids in 3D culture indicates a collective migration property with modulating high elastic modulus, suggesting activation for metastasis progress. This technology will be useful as a new class of 3D culture for cells and organoids due to its facility for deep-live observation and mechanical stiffness adjustment.
Collapse
Affiliation(s)
- Tomoyuki Suezawa
- Division of Applied Chemistry, Graduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Naoko Sasaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Yuichi Yukawa
- Division of Applied Chemistry, Graduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Nazgul Assan
- Division of Applied Chemistry, Graduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Yuta Uetake
- Division of Applied Chemistry, Graduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
- Innovative Catalysis Science Division, Institute for Open and Transdisciplinary Research Initiatives (ICS‐OTRI)Osaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Kunishige Onuma
- Department of Clinical Bio‐resource Research and DevelopmentKyoto University Graduate School of MedicineKyoto606–8304Japan
| | - Rino Kamada
- Division of Applied Chemistry, Graduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Daisuke Tomioka
- Division of Applied Chemistry, Graduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Hidehiro Sakurai
- Division of Applied Chemistry, Graduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
- Innovative Catalysis Science Division, Institute for Open and Transdisciplinary Research Initiatives (ICS‐OTRI)Osaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy CenterJapanese Foundation for Cancer ResearchTokyo135‐8550Japan
| | - Masahiro Inoue
- Department of Clinical Bio‐resource Research and DevelopmentKyoto University Graduate School of MedicineKyoto606–8304Japan
| | - Michiya Matsusaki
- Division of Applied Chemistry, Graduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| |
Collapse
|
31
|
Coppo R, Kondo J, Onuma K, Inoue M. Tracking the growth fate of single cells and isolating slow-growing cells in human colorectal cancer organoids. STAR Protoc 2023; 4:102395. [PMID: 37384521 PMCID: PMC10511865 DOI: 10.1016/j.xpro.2023.102395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/02/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023] Open
Abstract
Patient-derived tumor organoids are three-dimensionally cultured cancer cells that enable a suitable platform for studying heterogeneity and plasticity of cancer. We present a protocol for tracking the growth fate of single cells and isolating slow-growing cells in human colorectal cancer organoids. We describe steps for organoid preparation and culturing using the cancer-tissue-originating spheroid method, maintaining cell-cell contact throughout. We then detail a single-cell-derived spheroid-forming and growth assay, confirming single-cell plating, monitoring growth over time, and isolating slow-growing cells. For complete details on the use and execution of this protocol, please refer to Coppo et al.1.
Collapse
Affiliation(s)
- Roberto Coppo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Jumpei Kondo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kunishige Onuma
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
32
|
Shimizu S, Kondo J, Onuma K, Coppo R, Ota K, Kamada M, Harada Y, Tanaka Y, Nakazawa MA, Tamada Y, Okuno Y, Kawada K, Obama K, Coffey RJ, Fujiwara Y, Inoue M. Inhibition of the bone morphogenetic protein pathway suppresses tumor growth through downregulation of epidermal growth factor receptor in MEK/ERK-dependent colorectal cancer. Cancer Sci 2023; 114:3636-3648. [PMID: 37357017 PMCID: PMC10475764 DOI: 10.1111/cas.15882] [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: 11/10/2022] [Revised: 05/22/2023] [Accepted: 05/27/2023] [Indexed: 06/27/2023] Open
Abstract
The bone morphogenetic protein (BMP) pathway promotes differentiation and induces apoptosis in normal colorectal epithelial cells. However, its role in colorectal cancer (CRC) is controversial, where it can act as context-dependent tumor promoter or tumor suppressor. Here we have found that CRC cells reside in a BMP-rich environment based on curation of two publicly available RNA-sequencing databases. Suppression of BMP using a specific BMP inhibitor, LDN193189, suppresses the growth of select CRC organoids. Colorectal cancer organoids treated with LDN193189 showed a decrease in epidermal growth factor receptor, which was mediated by protein degradation induced by leucine-rich repeats and immunoglobulin-like domains protein 1 (LRIG1) expression. Among 18 molecularly characterized CRC organoids, suppression of growth by BMP inhibition correlated with induction of LRIG1 gene expression. Notably, knockdown of LRIG1 in organoids diminished the growth-suppressive effect of LDN193189. Furthermore, in CRC organoids, which are susceptible to growth suppression by LDN193189, simultaneous treatment with LDN193189 and trametinib, an FDA-approved MEK inhibitor, resulted in cooperative growth inhibition both in vitro and in vivo. Taken together, the simultaneous inhibition of BMP and MEK could be a novel treatment option in CRC cases, and evaluating in vitro growth suppression and LRIG1 induction by BMP inhibition using patient-derived organoids could offer functional biomarkers for predicting potential responders to this regimen.
Collapse
Affiliation(s)
- Shota Shimizu
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of MedicineTottori University Faculty of MedicineTottoriJapan
| | - Jumpei Kondo
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
- Department of BiochemistryOsaka International Cancer InstituteOsakaJapan
- Department of Molecular Biochemistry and Clinical Investigation, Division of Health ScienceOsaka University Graduate School of MedicineOsakaJapan
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Kunishige Onuma
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
| | - Roberto Coppo
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
| | - Kasumi Ota
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
| | - Mayumi Kamada
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
| | - Yohei Harada
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
| | - Yoshihisa Tanaka
- Graduate School of Pharmaceutical SciencesKyoto UniversityKyotoJapan
- RIKEN Center for Computational Science(R‐CCS)HPC/HPC‐ and AI‐driven Drug Development Platform DivisionKobeJapan
| | - Mai Adachi Nakazawa
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
- Department of Medical Data Intelligence and Data Analysis Division, Innovation Center for Health PromotionHirosaki UniversityHirosakiJapan
| | - Yoshinori Tamada
- Department of Medical Data Intelligence and Data Analysis Division, Innovation Center for Health PromotionHirosaki UniversityHirosakiJapan
| | - Yasushi Okuno
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
- RIKEN Center for Computational Science(R‐CCS)HPC/HPC‐ and AI‐driven Drug Development Platform DivisionKobeJapan
| | - Kenji Kawada
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Robert J. Coffey
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Yoshiyuki Fujiwara
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of MedicineTottori University Faculty of MedicineTottoriJapan
| | - Masahiro Inoue
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
- Department of BiochemistryOsaka International Cancer InstituteOsakaJapan
| |
Collapse
|
33
|
Anderle N, Schäfer-Ruoff F, Staebler A, Kersten N, Koch A, Önder C, Keller AL, Liebscher S, Hartkopf A, Hahn M, Templin M, Brucker SY, Schenke-Layland K, Schmees C. Breast cancer patient-derived microtumors resemble tumor heterogeneity and enable protein-based stratification and functional validation of individualized drug treatment. J Exp Clin Cancer Res 2023; 42:210. [PMID: 37596623 PMCID: PMC10436441 DOI: 10.1186/s13046-023-02782-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/28/2023] [Indexed: 08/20/2023] Open
Abstract
Despite tremendous progress in deciphering breast cancer at the genomic level, the pronounced intra- and intertumoral heterogeneity remains a major obstacle to the advancement of novel and more effective treatment approaches. Frequent treatment failure and the development of treatment resistance highlight the need for patient-derived tumor models that reflect the individual tumors of breast cancer patients and allow a comprehensive analyses and parallel functional validation of individualized and therapeutically targetable vulnerabilities in protein signal transduction pathways. Here, we introduce the generation and application of breast cancer patient-derived 3D microtumors (BC-PDMs). Residual fresh tumor tissue specimens were collected from n = 102 patients diagnosed with breast cancer and subjected to BC-PDM isolation. BC-PDMs retained histopathological characteristics, and extracellular matrix (ECM) components together with key protein signaling pathway signatures of the corresponding primary tumor tissue. Accordingly, BC-PDMs reflect the inter- and intratumoral heterogeneity of breast cancer and its key signal transduction properties. DigiWest®-based protein expression profiling of identified treatment responder and non-responder BC-PDMs enabled the identification of potential resistance and sensitivity markers of individual drug treatments, including markers previously associated with treatment response and yet undescribed proteins. The combination of individualized drug testing with comprehensive protein profiling analyses of BC-PDMs may provide a valuable complement for personalized treatment stratification and response prediction for breast cancer.
Collapse
Affiliation(s)
- Nicole Anderle
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany.
| | - Felix Schäfer-Ruoff
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
| | - Annette Staebler
- Institute of Pathology and Neuropathology, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Nicolas Kersten
- Interfaculty Institute for Bioinformatics and Medical Informatics (IBMI), Eberhard Karls University Tuebingen, Tuebingen, 72076, Germany
- FZI Research Center for Information Technology, 76131, Karlsruhe, Germany
| | - André Koch
- Department of Women's Health, University Women's Hospital, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Cansu Önder
- Department of Women's Health, University Women's Hospital, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Anna-Lena Keller
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
| | - Simone Liebscher
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Andreas Hartkopf
- Department of Women's Health, University Women's Hospital, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
- Department of Gynecology and Obstetrics, University Hospital of Ulm, 89081, Ulm, Germany
| | - Markus Hahn
- Department of Women's Health, University Women's Hospital, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Markus Templin
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
| | - Sara Y Brucker
- Department of Women's Health, University Women's Hospital, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Katja Schenke-Layland
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Christian Schmees
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany.
| |
Collapse
|
34
|
Qi H, Tan X, Zhang W, Zhou Y, Chen S, Zha D, Wang S, Wen J. The applications and techniques of organoids in head and neck cancer therapy. Front Oncol 2023; 13:1191614. [PMID: 37427120 PMCID: PMC10328716 DOI: 10.3389/fonc.2023.1191614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023] Open
Abstract
Head and neck cancer (HNC) is one of the most common cancers on the planet, with approximately 600,000 new cases diagnosed and 300,000 deaths every year. Research into the biological basis of HNC has advanced slowly over the past decades, which has made it difficult to develop new, more effective treatments. The patient-derived organoids (PDOs) are made from patient tumor cells, resembling the features of their tumors, which are high-fidelity models for studying cancer biology and designing new precision medicine therapies. In recent years, considerable effort has been focused on improving "organoids" technologies and identifying tumor-specific medicine using head and neck samples and a variety of organoids. A review of improved techniques and conclusions reported in publications describing the application of these techniques to HNC organoids is presented here. Additionally, we discuss the potential application of organoids in head and neck cancer research as well as the limitations associated with these models. As a result of the integration of organoid models into future precision medicine research and therapeutic profiling programs, the use of organoids will be extremely significant in the future.
Collapse
Affiliation(s)
- Hao Qi
- The Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Xiaolin Tan
- The Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
- Department of Clinical Nutrition, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Wenshuo Zhang
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yihong Zhou
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Shaoyi Chen
- The Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Dasong Zha
- The Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Siyang Wang
- The Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Jinming Wen
- The Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| |
Collapse
|
35
|
Roohani S, Loskutov J, Heufelder J, Ehret F, Wedeken L, Regenbrecht M, Sauer R, Zips D, Denker A, Joussen AM, Regenbrecht CRA, Kaul D. Photon and Proton irradiation in Patient-derived, Three-Dimensional Soft Tissue Sarcoma Models. BMC Cancer 2023; 23:577. [PMID: 37349697 DOI: 10.1186/s12885-023-11013-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 05/25/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Despite their heterogeneity, the current standard preoperative radiotherapy regimen for localized high-grade soft tissue sarcoma (STS) follows a one fits all approach for all STS subtypes. Sarcoma patient-derived three-dimensional cell culture models represent an innovative tool to overcome challenges in clinical research enabling reproducible subtype-specific research on STS. In this pilot study, we present our methodology and preliminary results using STS patient-derived 3D cell cultures that were exposed to different doses of photon and proton radiation. Our aim was: (i) to establish a reproducible method for irradiation of STS patient-derived 3D cell cultures and (ii) to explore the differences in tumor cell viability of two different STS subtypes exposed to increasing doses of photon and proton radiation at different time points. METHODS Two patient-derived cell cultures of untreated localized high-grade STS (an undifferentiated pleomorphic sarcoma (UPS) and a pleomorphic liposarcoma (PLS)) were exposed to a single fraction of photon or proton irradiation using doses of 0 Gy (sham irradiation), 2 Gy, 4 Gy, 8 Gy and 16 Gy. Cell viability was measured and compared to sham irradiation at two different time points (four and eight days after irradiation). RESULTS The proportion of viable tumor cells four days after photon irradiation for UPS vs. PLS were significantly different with 85% vs. 65% (4 Gy), 80% vs. 50% (8 Gy) and 70% vs. 35% (16 Gy). Proton irradiation led to similar diverging viability curves between UPS vs. PLS four days after irradiation with 90% vs. 75% (4 Gy), 85% vs. 45% (8 Gy) and 80% vs. 35% (16 Gy). Photon and proton radiation displayed only minor differences in cell-killing properties within each cell culture (UPS and PLS). The cell-killing effect of radiation sustained at eight days after irradiation in both cell cultures. CONCLUSIONS Pronounced differences in radiosensitivity are evident among UPS and PLS 3D patient-derived sarcoma cell cultures which may reflect the clinical heterogeneity. Photon and proton radiation showed similar dose-dependent cell-killing effectiveness in both 3D cell cultures. Patient-derived 3D STS cell cultures may represent a valuable tool to enable translational studies towards individualized subtype-specific radiotherapy in patients with STS.
Collapse
Affiliation(s)
- Siyer Roohani
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, Augustenburger Platz 1, 13353, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), 69120, Berlin, Heidelberg, Germany.
| | - Jürgen Loskutov
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Jens Heufelder
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, BerlinProtonen am Helmholtz-Zentrum Berlin, 14109, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Ophthalmology, 12200, Berlin, Germany
| | - Felix Ehret
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), 69120, Berlin, Heidelberg, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Lena Wedeken
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Manuela Regenbrecht
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125, Berlin, Germany
- Helios Klinikum Berlin-Buch, Schwanebecker Chaussee 50, 13125, Berlin, Germany
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Rica Sauer
- Institute of Pathology, Helios Klinikum Emil von Behring, Walterhöferstr. 11, 14165, Berlin, Germany
| | - Daniel Zips
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), 69120, Berlin, Heidelberg, Germany
| | - Andrea Denker
- Helmholtz-Zentrum Berlin für Materialien und Energie, 14109, Berlin, Germany
| | - Antonia M Joussen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Ophthalmology, 12200, Berlin, Germany
| | - Christian R A Regenbrecht
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125, Berlin, Germany
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125, Berlin, Germany
- Institut für Pathologie, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - David Kaul
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), 69120, Berlin, Heidelberg, Germany
| |
Collapse
|
36
|
Ando K, Suenaga Y, Kamijo T. DNA Ligase 4 Contributes to Cell Proliferation against DNA-PK Inhibition in MYCN-Amplified Neuroblastoma IMR32 Cells. Int J Mol Sci 2023; 24:ijms24109012. [PMID: 37240360 DOI: 10.3390/ijms24109012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/12/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Identifying the vulnerability of altered DNA repair machinery that displays synthetic lethality with MYCN amplification is a therapeutic rationale in unfavourable neuroblastoma. However, none of the inhibitors for DNA repair proteins are established as standard therapy in neuroblastoma. Here, we investigated whether DNA-PK inhibitor (DNA-PKi) could inhibit the proliferation of spheroids derived from neuroblastomas of MYCN transgenic mice and MYCN-amplified neuroblastoma cell lines. DNA-PKi exhibited an inhibitory effect on the proliferation of MYCN-driven neuroblastoma spheroids, whereas variable sensitivity was observed in those cell lines. Among them, the accelerated proliferation of IMR32 cells was dependent on DNA ligase 4 (LIG4), which comprises the canonical non-homologous end-joining pathway of DNA repair. Notably, LIG4 was identified as one of the worst prognostic factors in patients with MYCN-amplified neuroblastomas. It may play complementary roles in DNA-PK deficiency, suggesting the therapeutic potential of LIG4 inhibition in combination with DNA-PKi for MYCN-amplified neuroblastomas to overcome resistance to multimodal therapy.
Collapse
Affiliation(s)
- Kiyohiro Ando
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama 362-0806, Japan
| | - Yusuke Suenaga
- Chiba Cancer Center Research Institute, Chiba 260-8717, Japan
| | - Takehiko Kamijo
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama 362-0806, Japan
| |
Collapse
|
37
|
Nashimoto Y, Shishido S, Onuma K, Ino K, Inoue M, Shiku H. Oxygen metabolism analysis of a single organoid for non-invasive discrimination of cancer subpopulations with different growth capabilities. Front Bioeng Biotechnol 2023; 11:1184325. [PMID: 37274161 PMCID: PMC10232988 DOI: 10.3389/fbioe.2023.1184325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/08/2023] [Indexed: 06/06/2023] Open
Abstract
Heterogeneous nature is a pivotal aspect of cancer, rendering treatment problematic and frequently resulting in recurrence. Therefore, advanced techniques for identifying subpopulations of a tumour in an intact state are essential to develop novel screening platforms that can reveal differences in treatment response among subpopulations. Herein, we conducted a non-invasive analysis of oxygen metabolism on multiple subpopulations of patient-derived organoids, examining its potential utility for non-destructive identification of subpopulations. We utilised scanning electrochemical microscopy (SECM) for non-invasive analysis of oxygen metabolism. As models of tumours with heterogeneous subpopulations, we used patient-derived cancer organoids with a distinct growth potential established using the cancer tissue-originated spheroid methodology. Scanning electrochemical microscopy measurements enabled the analysis of the oxygen consumption rate (OCR) for individual organoids as small as 100 µm in diameter and could detect the heterogeneity amongst studied subpopulations, which was not observed in conventional colorectal cancer cell lines. Furthermore, our oxygen metabolism analysis of pre-isolated subpopulations with a slow growth potential revealed that oxygen consumption rate may reflect differences in the growth rate of organoids. Although the proposed technique currently lacks single-cell level sensitivity, the variability of oxygen metabolism across tumour subpopulations is expected to serve as an important indicator for the discrimination of tumour subpopulations and construction of novel drug screening platforms in the future.
Collapse
Affiliation(s)
- Yuji Nashimoto
- Institute of Bioengineering and Biomaterials (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Sendai, Miyagi, Japan
- Graduate School of Environmental Studies, Tohoku University, Sendai, Miyagi, Japan
- Graduate School of Engineering, Tohoku University, Sendai, Miyagi, Japan
| | - Shotaro Shishido
- Graduate School of Environmental Studies, Tohoku University, Sendai, Miyagi, Japan
| | | | - Kosuke Ino
- Graduate School of Engineering, Tohoku University, Sendai, Miyagi, Japan
| | - Masahiro Inoue
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hitoshi Shiku
- Graduate School of Environmental Studies, Tohoku University, Sendai, Miyagi, Japan
- Graduate School of Engineering, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
38
|
Goel R, Gulwani D, Upadhyay P, Sarangthem V, Singh TD. Unsung versatility of elastin-like polypeptide inspired spheroid fabrication: A review. Int J Biol Macromol 2023; 234:123664. [PMID: 36791934 DOI: 10.1016/j.ijbiomac.2023.123664] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/23/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023]
Abstract
Lately, 3D cell culture technique has gained a lot of appreciation as a research model. Augmented with technological advancements, the area of 3D cell culture is growing rapidly with a diverse array of scaffolds being tested. This is especially the case for spheroid cultures. The culture of cells as spheroids provides opportunities for unanticipated vision into biological phenomena with its application to drug discovery, metabolic profiling, stem cell research as well as tumor, and disease biology. Spheroid fabrication techniques are broadly categorised into matrix-dependent and matrix-independent techniques. While there is a profusion of spheroid fabrication substrates with substantial biological relevance, an economical, modular, and bio-compatible substrate for high throughput production of spheroids is lacking. In this review, we posit the prospects of elastin-like polypeptides (ELPs) as a broad-spectrum spheroid fabrication platform. Elastin-like polypeptides are nature inspired, size-tunable genetically engineered polymers with wide applicability in various arena of biological considerations, has been employed for spheroid culture with profound utility. The technology offers a cheap, high-throughput, reproducible alternative for spheroid culture with exquisite adaptability. Here, we will brief the applicability of 3D cultures as compared to 2D cultures with spheroids being the focal point of the review. Common approaches to spheroid fabrication are discussed with existential limitations. Finally, the versatility of elastin-like polypeptide inspired substrates for spheroid culture has been discussed.
Collapse
Affiliation(s)
- Ridhima Goel
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Deepak Gulwani
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Priyanka Upadhyay
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Vijaya Sarangthem
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Thoudam Debraj Singh
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
39
|
Parikh AS, Yu VX, Flashner S, Okolo OB, Lu C, Henick BS, Momen-Heravi F, Puram SV, Teknos T, Pan Q, Nakagawa H. Patient-derived three-dimensional culture techniques model tumor heterogeneity in head and neck cancer. Oral Oncol 2023; 138:106330. [PMID: 36773387 PMCID: PMC10126876 DOI: 10.1016/j.oraloncology.2023.106330] [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: 09/14/2022] [Revised: 12/08/2022] [Accepted: 01/25/2023] [Indexed: 02/11/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) outcomes remain stagnant, in part due to a poor understanding of HNSCC biology. The importance of tumor heterogeneity as an independent predictor of outcomes and treatment failure in HNSCC has recently come to light. With this understanding, 3D culture systems, including patient derived organoids (PDO) and organotypic culture (OTC), that capture this heterogeneity may allow for modeling and manipulation of critical subpopulations, such as p-EMT, as well as interactions between cancer cells and immune and stromal cells in the microenvironment. Here, we review work that has been done using PDO and OTC models of HNSCC, which demonstrates that these 3D culture models capture in vivo tumor heterogeneity and can be used to model tumor biology and treatment response in a way that faithfully recapitulates in vivo characteristics. As such, in vitro 3D culture models represent an important bridge between 2D monolayer culture and in vivo models such as patient derived xenografts.
Collapse
Affiliation(s)
- Anuraag S Parikh
- Department of Otolaryngology-Head and Neck Surgery, Columbia University, New York, NY, United States; Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Victoria X Yu
- Department of Otolaryngology-Head and Neck Surgery, Columbia University, New York, NY, United States
| | - Samuel Flashner
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, United States
| | - Ogoegbunam B Okolo
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Chao Lu
- Department of Genetics and Development, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, United States
| | - Brian S Henick
- Division of Hematology/Oncology, Department of Medicine, Columbia Unversity, New York, NY, United States
| | - Fatemeh Momen-Heravi
- Columbia University College of Dental Medicine, Columbia University, New York, NY, United States
| | - Sidharth V Puram
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO, United States; Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Theodoros Teknos
- Department of Otolaryngology, Case Western Reserve University, Cleveland, OH, United States
| | - Quintin Pan
- Department of Otolaryngology, Case Western Reserve University, Cleveland, OH, United States
| | - Hiroshi Nakagawa
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, United States.
| |
Collapse
|
40
|
Ex vivo chemosensitivity assay using primary ovarian cancer organoids for predicting clinical response and screening effective drugs. Hum Cell 2023; 36:752-761. [PMID: 36474106 DOI: 10.1007/s13577-022-00827-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
Selecting the best treatment for individual patients with cancer has attracted attention for improving clinical outcomes. Recent progress in organoid culture may lead to the development of personalized medicine. Unlike molecular-targeting drugs, there are no predictive methods for patient response to standard chemotherapies for ovarian cancer. We prepared organoids using the cancer tissue-originated spheroid (CTOS) method from 61 patients with ovarian cancer with 100% success rate. Chemosensitivity assays for paclitaxel and carboplatin were performed with 84% success rate using the primary organoids from 50 patients who received the chemotherapy. A wide range of sensitivities was observed among organoids for both drugs. All four clinically resistant organoids were resistant to both drugs in 18 cases in which clinical response information was available. Five out of 18 cases (28%) were double-resistant, the response rate of which was compatible with the clinical remission rate. Carboplatin was significantly more sensitive in serous than in clear cell subtypes (P = 0.025). We generated two lines of organoids, screened 1135 drugs, and found several drugs with better combinatory effects with carboplatin than with paclitaxel. Some drugs, including afatinib, have shown an additive effect with carboplatin. The organoid sensitivity assay did not predict the clinical outcomes, both progression free and overall survival.
Collapse
|
41
|
Chen L, Wei X, Gu D, Xu Y, Zhou H. Human liver cancer organoids: Biological applications, current challenges, and prospects in hepatoma therapy. Cancer Lett 2023; 555:216048. [PMID: 36603689 DOI: 10.1016/j.canlet.2022.216048] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023]
Abstract
Liver cancer and disease are among the most socially challenging global health concerns. Although organ transplantation, surgical resection and anticancer drugs are the main methods for the treatment of liver cancer, there are still no proven cures owing to the lack of donor livers and tumor heterogeneity. Recently, advances in tumor organoid technology have attracted considerable attention as they can simulate the spatial constructs and pathophysiological characteristics of tumorigenesis and metastasis in a more realistic manner. Organoids may further contribute to the development of tailored therapies. Combining organoids with other emerging techniques, such as CRISPR-HOT, organ-on-a-chip, and 3D bioprinting, may further develop organoids and address their bottlenecks to create more practical models that generalize different tissue or organ interactions in tumor progression. In this review, we summarize the various methods in which liver organoids may be generated and describe their biological and clinical applications, present challenges, and prospects for their integration with emerging technologies. These rapidly developing liver organoids may become the focus of in vitro clinical model development and therapeutic research for liver diseases in the near future.
Collapse
Affiliation(s)
- Lichan Chen
- Department of Laboratory Medicine, Inst Translat Med, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Xiafei Wei
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China.
| | - Dayong Gu
- Department of Laboratory Medicine, Inst Translat Med, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Yong Xu
- Department of Laboratory Medicine, Inst Translat Med, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Hongzhong Zhou
- Department of Laboratory Medicine, Inst Translat Med, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
42
|
Head and neck cancer patient-derived tumouroid cultures: opportunities and challenges. Br J Cancer 2023; 128:1807-1818. [PMID: 36765173 PMCID: PMC10147637 DOI: 10.1038/s41416-023-02167-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 02/12/2023] Open
Abstract
Head and neck cancers (HNC) are the seventh most prevalent cancer type globally. Despite their common categorisation, HNCs are a heterogeneous group of malignancies arising in various anatomical sites within the head and neck region. These cancers exhibit different clinical and biological manifestations, and this heterogeneity also contributes to the high rates of treatment failure and mortality. To evaluate patients who will respond to a particular treatment, there is a need to develop in vitro model systems that replicate in vivo tumour status. Among the methods developed, patient-derived cancer organoids, also known as tumouroids, recapitulate in vivo tumour characteristics including tumour architecture. Tumouroids have been used for general disease modelling and genetic instability studies in pan-cancer research. However, a limited number of studies have thus far been conducted using tumouroid-based drug screening. Studies have concluded that tumouroids can play an essential role in bringing precision medicine for highly heterogenous cancer types such as HNC.
Collapse
|
43
|
Harada Y, Sato A, Nakamura H, Kai K, Kitamura S, Nakamura T, Kurihara Y, Ikeda S, Sueoka E, Kimura S, Sueoka-Aragane N. Anti-cancer effect of afatinib, dual inhibitor of HER2 and EGFR, on novel mutation HER2 E401G in models of patient-derived cancer. BMC Cancer 2023; 23:77. [PMID: 36690964 PMCID: PMC9872313 DOI: 10.1186/s12885-022-10428-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/08/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Precision medicine with gene panel testing based on next-generation sequencing for patients with cancer is being used increasingly in clinical practice. HER2, which encodes the human epidermal growth factor receptor 2 (HER2), is a potentially important driver gene. However, therapeutic strategies aimed at mutations in the HER2 extracellular domain have not been clarified. We therefore investigated the effect of EGFR co-targeted therapy with HER2 on patient-derived cancer models with the HER2 extracellular domain mutation E401G, based on our previous findings that this mutation has an epidermal growth factor receptor (EGFR)-mediated activation mechanism. METHODS We generated a xenograft (PDX) and a cancer tissue-originated spheroid (CTOS) from a patient's cancer containing an amplified HER2 E401G mutation. With these platforms, we compared the efficacy of afatinib, a tyrosine kinase inhibitor having anti-HER2 and anti-EGFR activity, with two other therapeutic options: lapatinib, which has similar properties but weaker EGFR inhibition, and trastuzumab plus pertuzumab, for which evidence exists of treatment efficacy against cancers with wild-type HER2 amplification. Similar experiments were also performed with H2170, a cell line with wild-type HER2 amplification, to contrast the characteristics of these drug's efficacies against HER2 E401G. RESULTS We confirmed that PDX and CTOS retained morphological and immunohistochemical characteristics and HER2 gene profiles of the original tumor. In both PDX and CTOS, afatinib reduced tumor size more than lapatinib or trastuzumab plus pertuzumab. In addition, afatinib treatment resulted in a statistically significant reduction in HER2 copy number at the end of treatment. On the other hand, in H2170 xenografts with wild-type HER2 amplification, trastuzumab plus pertuzumab was most effective. CONCLUSIONS Afatinib, a dual inhibitor of HER2 and EGFR, showed a promising effect on cancers with amplified HER2 E401G, which have an EGFR-mediated activation mechanism. Analysis of the activation mechanisms of mutations and development of therapeutic strategies based on those mechanisms are critical in precision medicine for cancer patients.
Collapse
Affiliation(s)
- Yohei Harada
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
- Graduate School of Medicine, Kyoto University, 53 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akemi Sato
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Hideaki Nakamura
- Department of Transfusion Medicine, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Keita Kai
- Department of Pathology, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Sho Kitamura
- Department of Pathology, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Tomomi Nakamura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Eisaburo Sueoka
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan.
| |
Collapse
|
44
|
Coppo R, Kondo J, Iida K, Okada M, Onuma K, Tanaka Y, Kamada M, Ohue M, Kawada K, Obama K, Inoue M. Distinct but interchangeable subpopulations of colorectal cancer cells with different growth fates and drug sensitivity. iScience 2023; 26:105962. [PMID: 36718360 PMCID: PMC9883198 DOI: 10.1016/j.isci.2023.105962] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/22/2022] [Accepted: 01/09/2023] [Indexed: 01/14/2023] Open
Abstract
Dynamic changes in cell properties lead to intratumor heterogeneity; however, the mechanisms of nongenetic cellular plasticity remain elusive. When the fate of each cell from colorectal cancer organoids was tracked through a clonogenic growth assay, the cells showed a wide range of growth ability even within the clonal organoids, consisting of distinct subpopulations; the cells generating large spheroids and the cells generating small spheroids. The cells from the small spheroids generated only small spheroids (S-pattern), while the cells from the large spheroids generated both small and large spheroids (D-pattern), both of which were tumorigenic. Transition from the S-pattern to the D-pattern occurred by various extrinsic triggers, in which Notch signaling and Musashi-1 played a key role. The S-pattern spheroids were resistant to chemotherapy and transited to the D-pattern upon drug treatment through Notch signaling. As the transition is linked to the drug resistance, it can be a therapeutic target.
Collapse
Affiliation(s)
- Roberto Coppo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jumpei Kondo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keita Iida
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Mariko Okada
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Kunishige Onuma
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihisa Tanaka
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan,RIKEN Center for Computational Science, HPC- and AI-driven Drug Development Platform Division, Biomedical Computational Intelligence Unit, Hyogo, Japan
| | - Mayumi Kamada
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masayuki Ohue
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Kenji Kawada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan,Corresponding author
| |
Collapse
|
45
|
Geometric tumor embolic budding characterizes inflammatory breast cancer. Breast Cancer Res Treat 2023; 197:461-478. [PMID: 36473978 PMCID: PMC9734724 DOI: 10.1007/s10549-022-06819-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE Inflammatory breast cancer (IBC) is characterized by numerous tumor emboli especially within dermal lymphatics. The explanation remains a mystery. METHODS This study combines experimental studies with two different IBC xenografts with image algorithmic studies utilizing human tissue microarrays (TMAs) of IBC vs non-IBC cases to support a novel hypothesis to explain IBC's sina qua non signature of florid lymphovascular emboli. RESULTS In the human TMAs, compared to tumor features like nuclear grade (size), mitosis and Ki-67 immunoreactivity which show that IBC is only modestly more proliferative with larger nuclei than non-IBC, what really sets IBC apart is the markedly greater number of tumor emboli and distinctly smaller emboli whose numbers indicate geometric or exponential differences between IBC and non-IBC. In the experimental xenograft studies, Mary-X gives rise to tight spheroids in vitro which exhibit dynamic budding into smaller daughter spheroids whereas Karen-X exhibits only loose non-budding aggregates. Furthermore Mary-X emboli also bud dramatically into smaller daughter emboli in vivo. The mechanism that regulates this involves the generation of E-cad/NTF1, a calpain-mediated cleavage 100 kDa product of 120 kDa full length membrane E-cadherin. Inhibiting this calpain-mediated cleavage of E-cadherin by blocking either the calpain site of cleavage (SC) or the site of binding (SB) with specific decapeptides that both penetrate the cell membrane and mimic either the cleavage site or the binding site on E-cadherin, inhibits the generation of E-cad/NTF1 in a dose-dependent manner, reduces spheroid compactness and decreases budding. CONCLUSION Since E-cad/NFT1 retains the p120ctn binding site but loses the α-and β-catenin sites, promoting its 360° distribution around the cell's membrane, the vacilating levels of this molecule trigger budding of both the spheroids as well as the emboli. Recurrent and geometric budding of parental emboli into daughter emboli then would account for the plethora of emboli seen in IBC.
Collapse
|
46
|
Nashimoto Y, Mukomoto R, Imaizumi T, Terai T, Shishido S, Ino K, Yokokawa R, Miura T, Onuma K, Inoue M, Shiku H. Electrochemical sensing of oxygen metabolism for a three-dimensional cultured model with biomimetic vascular flow. Biosens Bioelectron 2023; 219:114808. [PMID: 36327566 DOI: 10.1016/j.bios.2022.114808] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
Microphysiological systems (MPSs) with three-dimensional (3D) cultured models have attracted considerable interest because of their potential to mimic human health and disease conditions. Recent MPSs have shown significant advancements in engineering perfusable vascular networks integrated with 3D culture models, realizing a more physiological environment in vitro; however, a sensing system that can monitor their activity under biomimetic vascular flow is lacking. We designed an open-top microfluidic device with sensor capabilities and demonstrated its application in analyzing oxygen metabolism in vascularized 3D tissue models. We first validated the platform by using human lung fibroblast (hLF) spheroids. Then, we applied the platform to a patient-derived cancer organoid and evaluated the changes in oxygen metabolism during drug administration through the vascular network. We found that the platform could integrate a perfusable vascular network with 3D cultured cells, and the electrochemical sensor could detect the change in oxygen metabolism in a quantitative, non-invasive, and real-time manner. This platform would become a monitoring system for 3D cultured cells integrated with a perfusable vascular network.
Collapse
Affiliation(s)
- Yuji Nashimoto
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Miyagi, 980-8578, Japan; Graduate School of Engineering, Tohoku University, Miyagi, 980-8579, Japan; Graduate School of Environmental Studies, Tohoku University, Miyagi, 980-8579, Japan.
| | - Rei Mukomoto
- Graduate School of Environmental Studies, Tohoku University, Miyagi, 980-8579, Japan
| | - Takuto Imaizumi
- Graduate School of Environmental Studies, Tohoku University, Miyagi, 980-8579, Japan
| | - Takato Terai
- Graduate School of Environmental Studies, Tohoku University, Miyagi, 980-8579, Japan
| | - Shotaro Shishido
- Graduate School of Environmental Studies, Tohoku University, Miyagi, 980-8579, Japan
| | - Kosuke Ino
- Graduate School of Engineering, Tohoku University, Miyagi, 980-8579, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Takashi Miura
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kunishige Onuma
- Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Masahiro Inoue
- Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Hitoshi Shiku
- Graduate School of Engineering, Tohoku University, Miyagi, 980-8579, Japan; Graduate School of Environmental Studies, Tohoku University, Miyagi, 980-8579, Japan.
| |
Collapse
|
47
|
Lopez‐Mejia IC, Pijuan J, Navaridas R, Santacana M, Gatius S, Velasco A, Castellà G, Panosa A, Cabiscol E, Pinyol M, Coll L, Bonifaci N, Peña LP, Vidal A, Villanueva A, Gari E, Llobet‐Navàs D, Fajas L, Matias‐Guiu X, Yeramian A. Oxidative stress-induced FAK activation contributes to uterine serous carcinoma aggressiveness. Mol Oncol 2022; 17:98-118. [PMID: 36409196 PMCID: PMC9812840 DOI: 10.1002/1878-0261.13346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/18/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022] Open
Abstract
Uterine serous carcinoma (USC) is an aggressive form of endometrial cancer (EC), characterized by its high propensity for metastases. In fact, while endometrioid endometrial carcinoma (EEC), which accounts for 85% of EC, presents a good prognosis, USC is the most frequently fatal. Herein, we used for the first time a peptide-based tyrosine-kinase-activity profiling approach to quantify the changes in tyrosine kinase activation between USC and EEC. Among the tyrosine kinases highly activated in USC, we identified focal adhesion kinase (FAK). We conducted mechanistic studies using cellular models. In a USC cell line, targeting FAK either by inhibitors PF-573228 and defactinib (VS-6063) or by gene silencing limits 3D cell growth and reduces cell migration. Moreover, results from our studies suggest that oxidative stress is increased in USC tumors compared to EEC ones. Reactive oxygen species (ROS) induce tyrosine phosphorylation of FAK and a concomitant tyrosine phosphorylation of paxillin, a mediator of FAK signal transduction. Mechanistically, by tracking hundreds of individual cells per condition, we show that ROS increased cell distance and migration velocity, highlighting the role of ROS-FAK-PAX signaling in cell migration. Both defactinib and ROS scavenger N-acetylcysteine (NAC) revert this effect, pointing toward ROS as potential culprits for the increase in USC cell motility. A proof of concept of the role of FAK in controlling cell growth was obtained in in vivo experiments using cancer-tissue-originated spheroids (CTOS) and a patient-derived orthotopic xenograft model (orthoxenograft/PDOX). Defactinib reduces cell proliferation and protein oxidation, supporting a pro-tumoral antioxidant role of FAK, whereas antioxidant NAC reverts FAK inhibitor effects. Overall, our data points to ROS-mediated FAK activation in USC as being responsible for the poor prognosis of this tumor type and emphasize the potential of FAK inhibition for USC treatment.
Collapse
Affiliation(s)
| | - Jordi Pijuan
- Laboratory of Neurogenetics and Molecular Medicine – Pediatric Institute of Rare DiseasesInstitut de Recerca Sant Joan de DéuBarcelonaSpain
| | - Raúl Navaridas
- Departament de Ciències Mèdiques Bàsiques, IRBLleidaUniversity of LleidaSpain
| | - Maria Santacana
- Pathology Group, Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova (HUAV), IRBLleidaUniversity of LleidaSpain
| | - Sònia Gatius
- Pathology Group, Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova (HUAV), IRBLleidaUniversity of LleidaSpain
| | - Ana Velasco
- Pathology Group, Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova (HUAV), IRBLleidaUniversity of LleidaSpain
| | - Gerard Castellà
- Biostatistics Unit, Hospital Universitari Arnau de Vilanova, IRB‐LleidaUniversity of LleidaSpain
| | - Anaïs Panosa
- Flow Cytometry and Confocal Microscopy Unit, IRBLleidaUniversity of LleidaSpain
| | - Elisa Cabiscol
- Departament de Ciències Mèdiques Bàsiques, IRBLleidaUniversity of LleidaSpain
| | - Miquel Pinyol
- Pathology Group, Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova (HUAV), IRBLleidaUniversity of LleidaSpain
| | - Laura Coll
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)Gran via de l'HospitaletBarcelonaSpain
| | - Núria Bonifaci
- Pathology Group, Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova (HUAV), IRBLleidaUniversity of LleidaSpain,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos III (ISCIII)MadridSpain
| | - Laura Plata Peña
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)Gran via de l'HospitaletBarcelonaSpain
| | - August Vidal
- Department of Pathology‐HospitalUniversitari de BellvitgeBarcelonaSpain,Xenopat S.L., Parc Cientific de Barcelona (PCB)Spain
| | - Alberto Villanueva
- Xenopat S.L., Parc Cientific de Barcelona (PCB)Spain,Program Against Cancer Therapeutic Resistance (ProCURE), ICO, IDIBELLBarcelonaSpain
| | - Eloi Gari
- Departament de Ciències Mèdiques Bàsiques, IRBLleidaUniversity of LleidaSpain
| | - David Llobet‐Navàs
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)Gran via de l'HospitaletBarcelonaSpain,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos III (ISCIII)MadridSpain
| | - Lluis Fajas
- Center for Integrative GenomicsUniversity of LausanneSwitzerland
| | - Xavier Matias‐Guiu
- Pathology Group, Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova (HUAV), IRBLleidaUniversity of LleidaSpain,Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)Gran via de l'HospitaletBarcelonaSpain,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos III (ISCIII)MadridSpain,Department of Pathology‐HospitalUniversitari de BellvitgeBarcelonaSpain
| | - Andrée Yeramian
- Pathology Group, Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova (HUAV), IRBLleidaUniversity of LleidaSpain
| |
Collapse
|
48
|
Tenschert E, Kern J, Affolter A, Rotter N, Lammert A. Optimisation of Conditions for the Formation of Spheroids of Head and Neck Squamous Cell Carcinoma Cell Lines for Use as Animal Alternatives. Altern Lab Anim 2022; 50:414-422. [PMID: 36263982 DOI: 10.1177/02611929221135042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The use of in vitro 3-D cell culture models in cancer research has yielded substantial gains in knowledge on various aspects of tumour biology. Such cell culture models could be useful in the study of head and neck squamous cell carcinoma (HNSCC), where mimicking intratumoral and intertumoral heterogeneity is especially challenging. Our research aims to establish 3-D spheroid models for HNSCC that reproduce in vitro the connections between tumour cells and the surrounding microenvironment. The aims of this study were to determine the optimal conditions for the culture and use of spheroids from HNSCC cell lines and optimal timepoint for using the spheroids obtained, to evaluate the effects of coculture with tumour-specific fibroblasts on spheroid formation, and to investigate spheroid responses to cisplatin treatment. Four HNSCC cell lines (UMSCC-11A, UMSCC-11B, UMSCC-22B and UD-SCC-01) were seeded in flat or round bottom well ultra-low attachment spheroid plates, and spheroid formation was evaluated. The HNSCC cell lines were then cocultured with stromal cells of the tumour microenvironment, producing an accelerated formation of dense spheroids. The viability of cells within the spheroids was assessed during cell culture by using a fluorescent dye. Our results suggest that: three out of the four cell lines tested could form usable spheroids with acceptable viability; the addition of stromal cells did not improve the number of viable cells; and the use of round bottom well plates supported the formation of a single spheroid, whereas flat bottom well plates led to the formation of multiple spheroids of different sizes.
Collapse
Affiliation(s)
- Esther Tenschert
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Johann Kern
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Annette Affolter
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Nicole Rotter
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Anne Lammert
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| |
Collapse
|
49
|
BDNF/TRKB axis provokes EMT progression to induce cell aggressiveness via crosstalk with cancer-associated fibroblasts in human parotid gland cancer. Sci Rep 2022; 12:17553. [PMID: 36266462 PMCID: PMC9584965 DOI: 10.1038/s41598-022-22377-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 10/13/2022] [Indexed: 01/13/2023] Open
Abstract
Parotid gland cancer (PGC) is a rare malignancy and its molecular characteristics remain poorly understood, which has precluded the development of effective drug therapies. Given the poor prognosis of many human cancers in which tropomyosin receptor kinase B (TRKB) is highly expressed, we investigated the involvement of brain-derived neurotrophic factor (BDNF)/TRKB pathway in PGC cells using clinical specimens and observed upregulation of TRKB and BDNF. In primary culture systems of patient-derived PGC cells and cancer-associated fibroblasts (CAFs), PGC cells co-cultured with CAFs exhibited significant upregulation of BDNF and epithelial-mesenchymal transition (EMT). Similar results were observed in PGC cells treated with conditioned medium from co-cultures of PGC cells with CAFs. Administration of TRK inhibitors suppressed BDNF-induced cell migration in PGC cells. Immunohistochemical and clinicopathological analyses of tumors from patients with PGC revealed that BDNF and TRKB were highly expressed in both tumor cells and stromal cells such as CAFs, and TRKB expression levels in PGC cells were significantly correlated with aggressive features, including vascular invasion, nodal metastasis, and poor prognosis. Collectively, these data suggest that the BDNF/TRKB pathway regulates PGC cell aggressiveness via crosstalk with CAFs and is a potential therapeutic target for PGC harboring invasive and metastatic features.
Collapse
|
50
|
Parihar A, Pandita V, Khan R. 3D printed human organoids: High throughput system for drug screening and testing in current COVID-19 pandemic. Biotechnol Bioeng 2022; 119:2669-2688. [PMID: 35765706 DOI: 10.1002/bit.28166] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/27/2022] [Accepted: 06/24/2022] [Indexed: 11/07/2022]
Abstract
In the current pandemic, scenario the world is facing a huge shortage of effective drugs and other prophylactic medicine to treat patients which created havoc in several countries with poor resources. With limited demand and supply of effective drugs, researchers rushed to repurpose the existing approved drugs for the treatment of COVID-19. The process of drug screening and testing is very costly and requires several steps for validation and treatment efficacy evaluation ranging from in-vitro to in-vivo setups. After these steps, a clinical trial is mandatory for the evaluation of treatment efficacy and side effects in humans. These processes enhance the overall cost and sometimes the lead molecule show adverse effects in humans and the trial ends up in the final stages. Recently with the advent of three-dimensional (3D) organoid culture which mimics the human tissue exactly the process of drug screening and testing can be done in a faster and cost-effective manner. Further 3D organoids prepared from stems cells taken from individuals can be beneficial for personalized drug therapy which could save millions of lives. This review discussed approaches and techniques for the synthesis of 3D-printed human organoids for drug screening. The key findings of the usage of organoids for personalized medicine for the treatment of COVID-19 have been discussed. In the end, the key challenges for the wide applicability of human organoids for drug screening with prospects of future orientation have been included.
Collapse
Affiliation(s)
- Arpana Parihar
- Industrial Waste Utilization, Nano and Biomaterials, CSIR-Advanced Materials and Processes Research Institute (AMPRI), Bhopal, Madhya Pradesh, India
| | - Vasundhara Pandita
- Department of Biosciences, Barkatullah University, Bhopal, Madhya Pradesh, India
| | - Raju Khan
- Industrial Waste Utilization, Nano and Biomaterials, CSIR-Advanced Materials and Processes Research Institute (AMPRI), Bhopal, Madhya Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|