1
|
Yang J, Hawthorne L, Stack S, Blagg B, Ali A, Zorlutuna P. Engineered Age-Mimetic Breast Cancer Models Reveal Differential Drug Responses in Young and Aged Microenvironments. Adv Healthc Mater 2025; 14:e2404461. [PMID: 39821643 PMCID: PMC11960616 DOI: 10.1002/adhm.202404461] [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: 12/02/2024] [Indexed: 01/19/2025]
Abstract
Aging is one of the most significant risk factors for breast cancer. With the growing interest in the alterations of the aging breast tissue microenvironment, it is identified that aging is related to tumorigenesis, invasion, and drug resistance. However, current pre-clinical disease models often neglect the impact of aging and sometimes result in worse clinical outcomes. In this study, aged animal-generated materials are utilized to create and validate a novel age-mimetic breast cancer model that generates an aging microenvironment for cells and alters cells toward a more invasive phenotype found in the aged environment. Furthermore, the age-mimetic models are utilized for 3D breast cancer invasion assessment and high-throughput screening of over 700 drugs in the FDA-approved drug library. 36 potential effective drug targets as well as 34 potential drug targets with different drug responses in different age groups are identified, demonstrating the potential of this age-mimetic breast cancer model for further in-depth breast cancer studies and drug development.
Collapse
Affiliation(s)
- Jun Yang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Lauren Hawthorne
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Sharon Stack
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| | - Brian Blagg
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| | - Aktar Ali
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| | - Pinar Zorlutuna
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| |
Collapse
|
2
|
Isinelli G, Failla S, Plebani R, Prete A. Exploring oncology treatment strategies with tyrosine kinase inhibitors through advanced 3D models (Review). MEDICINE INTERNATIONAL 2025; 5:13. [PMID: 39790707 PMCID: PMC11707505 DOI: 10.3892/mi.2024.212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025]
Abstract
The limitations of two-dimensional (2D) models in cancer research have hindered progress in fully understanding the complexities of drug resistance and therapeutic failures. However, three-dimensional (3D) models provide a more accurate representation of in vivo environments, capturing critical cellular interactions and dynamics that are essential in evaluating the efficacy and toxicity of tyrosine kinase inhibitors (TKIs). These advanced models enable researchers to explore drug resistance mechanisms with greater precision, optimizing treatment strategies and improving the predictive accuracy of clinical outcomes. By leveraging 3D models, it will be possible to deepen the current understanding of TKIs and drive forward innovations in cancer treatment. The present review discusses the limitations of 2D models and the transformative impact of 3D models on oncology research, highlighting their roles in addressing the challenges of 2D systems and advancing TKI studies.
Collapse
Affiliation(s)
- Giorgia Isinelli
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Chemistry, Biology and Biotechnology, University of Perugia, I-06123 Perugia, Italy
| | - Sharon Failla
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Roberto Plebani
- Department of Medical, Oral and Biotechnological Sciences, ‘G. D'Annunzio’ University, I-66100 Chieti-Pescara, Italy
| | - Alessandro Prete
- Department of Clinical and Experimental Medicine, Endocrine Unit 2, University of Pisa, I-56122 Pisa, Italy
| |
Collapse
|
3
|
Yang J, Hawthorne L, Stack S, Blagg B, Ali A, Zorlutuna P. Engineered Age-Mimetic Breast Cancer Models Reveal Differential Drug Responses in Young and Aged Microenvironments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.06.616903. [PMID: 39416111 PMCID: PMC11482747 DOI: 10.1101/2024.10.06.616903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Aging is one of the most significant risk factors for breast cancer. With the growing interests in the alterations of the aging breast tissue microenvironment, it has been identified that aging is related to tumorigenesis, invasion, and drug resistance. However, current pre-clinical disease models often neglect the impact of aging and sometimes result in worse clinical outcomes. In this study, we utilized aged animal-generated materials to create and validate a novel age-mimetic breast cancer model that generates an aging microenvironment for cells and alters cells towards a phenotype found in the aged environment. Furthermore, we utilized the age-mimetic models for 3D breast cancer invasion assessment and high-throughput screening of over 700 drugs in the FDA-approved drug library. We identified 36 potential effective drug targets and 34 potential drug targets with different drug responses in different age groups, demonstrating the potential of this age-mimetic breast cancer model for further in-depth breast cancer studies and drug development.
Collapse
Affiliation(s)
- Jun Yang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Lauren Hawthorne
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Sharon Stack
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| | - Brian Blagg
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| | - Aktar Ali
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| | - Pinar Zorlutuna
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| |
Collapse
|
4
|
Flörkemeier I, Antons LK, Weimer JP, Hedemann N, Rogmans C, Krüger S, Scherließ R, Dempfle A, Arnold N, Maass N, Bauerschlag DO. Multicellular ovarian cancer spheroids: novel 3D model to mimic tumour complexity. Sci Rep 2024; 14:23526. [PMID: 39384844 PMCID: PMC11464915 DOI: 10.1038/s41598-024-73680-6] [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: 09/19/2024] [Indexed: 10/11/2024] Open
Abstract
In vitro, spheroid models have become well established in cancer research because they can better mimic certain characteristics of in vivo tumours. However, interaction with the tumour microenvironment, such as cancer-associated fibroblasts, plays a key role in tumour progression. We initially focused on the interaction of tumour cells with fibroblasts. To model this interaction, we developed a spheroid model of ovarian cancer and fibroblasts. To this end, ovarian cancer cell lines and ex vivo primary cells were simultaneously and sequentially seeded with fibroblasts in a scaffold-free system at different ratios and subsequently characterized with respect to changes in morphology, proliferation, and viability. We demonstrated that co-cultures are able to form by far more compact spheroids, especially in cells that form aggregates in mono-culture. In addition, the co-cultures were able to increase proliferation and sensitivity to cisplatin. Simultaneous seeding led fibroblasts invade the core in both cell lines and primary cells. These results show differences in formation, firmness, and size between co-culture and mono-culture. Our model is designed to better represent and characterize the mutual influencing factors of fibroblasts and tumour cells. Fibroblast-supplemented multicellular spheroids are a valuable tool for tumour microenvironment interaction and new drug discovery.
Collapse
Affiliation(s)
- Inken Flörkemeier
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany.
- KiNSIS Priority Research Area, Kiel University, Kiel, Germany.
| | - Lisa K Antons
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Jörg P Weimer
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Nina Hedemann
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Christoph Rogmans
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Sandra Krüger
- Department of Pathology, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Kiel, Germany
- KiNSIS Priority Research Area, Kiel University, Kiel, Germany
| | - Astrid Dempfle
- Institute of Medical Informatics and Statistics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Norbert Arnold
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Nicolai Maass
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Dirk O Bauerschlag
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
- Department of Gynaecology, Jena University Hospital, Jena, Germany
| |
Collapse
|
5
|
Arora S, Singh S, Mittal A, Desai N, Khatri DK, Gugulothu D, Lather V, Pandita D, Vora LK. Spheroids in cancer research: Recent advances and opportunities. J Drug Deliv Sci Technol 2024; 100:106033. [DOI: 10.1016/j.jddst.2024.106033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024]
|
6
|
Kim O, Sergi Z, Yu G, Yamamoto K, Quezado M, Abdullaev Z, Crooks DR, Kishimoto S, Li Q, Lu P, Blackman B, Andresson T, Wu X, Tran B, Wei JS, Zhang W, Zhang M, Song H, Khan J, Krishna MC, Brender JR, Wu J. A patient-derived cell model for malignant transformation in IDH-mutant glioma. Acta Neuropathol Commun 2024; 12:148. [PMID: 39256867 PMCID: PMC11385154 DOI: 10.1186/s40478-024-01860-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/31/2024] [Indexed: 09/12/2024] Open
Abstract
Malignant transformation (MT) is commonly seen in IDH-mutant gliomas. There has been a growing research interest in revealing its underlying mechanisms and intervening prior to MT at the early stages of the transforming process. Here we established a unique pair of matched 3D cell models: 403L, derived from a low-grade glioma (LGG), and 403H, derived from a high-grade glioma (HGG), by utilizing IDH-mutant astrocytoma samples from the same patient when the tumor was diagnosed as WHO grade 2 (tumor mutational burden (TMB) of 3.96/Mb) and later as grade 4 (TMB of 70.07/Mb), respectively. Both cell models were authenticated to a patient's sample retaining endogenous expression of IDH1 R132H. DNA methylation profiles of the parental tumors referred to LGG and HGG IDH-mutant glioma clusters. The immunopositivity of SOX2, NESTIN, GFAP, OLIG2, and beta 3-Tubulin suggested the multilineage potential of both models. 403H was more prompt to cell invasion and developed infiltrative HGG in vivo. The differentially expressed genes (DEGs) from the RNA sequencing analysis revealed the tumor invasion and aggressiveness related genes exclusively upregulated in the 403H model. Pathway analysis showcased an enrichment of genes associated with epithelial-mesenchymal transition (EMT) and Notch signaling pathways in 403H and 403L, respectively. Mass spectrometry-based targeted metabolomics and hyperpolarized (HP) 1-13C pyruvate in-cell NMR analyses demonstrated significant alterations in the TCA cycle and fatty acid metabolism. Citrate, glutamine, and 2-HG levels were significantly higher in 403H. To our knowledge, this is the first report describing the development of a matched pair of 3D patient-derived cell models representative of MT and temozolomide (TMZ)-induced hypermutator phenotype (HMP) in IDH-mutant glioma, providing insights into genetic and metabolic changes during MT/HMP. This novel in vitro model allows further investigation of the mechanisms of MT at the cellular level.
Collapse
Affiliation(s)
- Olga Kim
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 1142A, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Zach Sergi
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 1142A, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Guangyang Yu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 1142A, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Kazutoshi Yamamoto
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Martha Quezado
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Zied Abdullaev
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Danel R Crooks
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Shun Kishimoto
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Qi Li
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 1142A, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Peng Lu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 1142A, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Burchelle Blackman
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, Rockville, MD, 20850, USA
| | - Thorkell Andresson
- Protein Characterization Laboratory, Leidos Biomedical Inc / Frederick National Laboratory for Cancer Research, Frederick, MD, 21701, USA
| | - Xiaolin Wu
- Genomics Technology Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21701, USA
| | - Bao Tran
- Sequencing Facility, Leidos Biomedical Inc / Frederick National Laboratory for Cancer Research, Frederick, MD, 21701, USA
| | - Jun S Wei
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wei Zhang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 1142A, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Meili Zhang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 1142A, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Hua Song
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 1142A, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Murali C Krishna
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jeffrey R Brender
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jing Wu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 1142A, 37 Convent Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
7
|
Wang G, Mao X, Wang W, Wang X, Li S, Wang Z. Bioprinted research models of urological malignancy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230126. [PMID: 39175884 PMCID: PMC11335473 DOI: 10.1002/exp.20230126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/08/2024] [Indexed: 08/24/2024]
Abstract
Urological malignancy (UM) is among the leading threats to health care worldwide. Recent years have seen much investment in fundamental UM research, including mechanistic investigation, early diagnosis, immunotherapy, and nanomedicine. However, the results are not fully satisfactory. Bioprinted research models (BRMs) with programmed spatial structures and functions can serve as powerful research tools and are likely to disrupt traditional UM research paradigms. Herein, a comprehensive review of BRMs of UM is presented. It begins with a brief introduction and comparison of existing UM research models, emphasizing the advantages of BRMs, such as modeling real tissues and organs. Six kinds of mainstream bioprinting techniques used to fabricate such BRMs are summarized with examples. Thereafter, research advances in the applications of UM BRMs, such as culturing tumor spheroids and organoids, modeling cancer metastasis, mimicking the tumor microenvironment, constructing organ chips for drug screening, and isolating circulating tumor cells, are comprehensively discussed. At the end of this review, current challenges and future development directions of BRMs and UM are highlighted from the perspective of interdisciplinary science.
Collapse
Affiliation(s)
- Guanyi Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| | - Xiongmin Mao
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Wang Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiaolong Wang
- Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Sheng Li
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Zijian Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| |
Collapse
|
8
|
Gayan S, Teli A, Sonawane A, Dey T. Impact of Chemotherapeutic Stress Depends on The Nature of Breast Cancer Spheroid and Induce Behavioral Plasticity to Resistant Population. Adv Biol (Weinh) 2024; 8:e2300271. [PMID: 38063815 DOI: 10.1002/adbi.202300271] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 11/20/2023] [Indexed: 04/15/2024]
Abstract
Cellular or tumor dormancy, identified recently as one of the main reasons behind post-therapy recurrence, can be caused by diverse reasons. Chemotherapy has recently been recognized as one of such reasons. However, in-depth studies of chemotherapy-induced dormancy are lacking due to the absence of an in vitro human-relevant model tailor-made for such a scenario. This report utilized multicellular breast cancer spheroid to create a primary platform for establishing a chemotherapy-induced dormancy model. It is observed that extreme chemotherapeutic stress affects invasive and non-invasive spheroids differently. Non-invasive spheroids exhibit more resilience and maintain viability and migrational ability, while invasive spheroids display heightened susceptibility and improved tumorigenic capacity. Heterogenous spheroids exhibit increased tumorigenic capacity while show minimal survival ability. Further probing of chemotherapeutically dormant spheroids is needed to understand the molecular mechanism and identify dormancy-related markers to achieve therapeutic success in the future.
Collapse
Affiliation(s)
- Sukanya Gayan
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Abhishek Teli
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Akshay Sonawane
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Tuli Dey
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| |
Collapse
|
9
|
Cai X, Refaat A, Gan PY, Fan B, Yu H, Thang SH, Drummond CJ, Voelcker NH, Tran N, Zhai J. Angiopep-2-Functionalized Lipid Cubosomes for Blood-Brain Barrier Crossing and Glioblastoma Treatment. ACS APPLIED MATERIALS & INTERFACES 2024; 16:12161-12174. [PMID: 38416873 DOI: 10.1021/acsami.3c14709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain cancer with high malignancy and resistance to conventional treatments, resulting in a bleak prognosis. Nanoparticles offer a way to cross the blood-brain barrier (BBB) and deliver precise therapies to tumor sites with reduced side effects. In this study, we developed angiopep-2 (Ang2)-functionalized lipid cubosomes loaded with cisplatin (CDDP) and temozolomide (TMZ) for crossing the BBB and providing targeted glioblastoma therapy. Developed lipid cubosomes showed a particle size of around 300 nm and possessed an internal ordered inverse primitive cubic phase, a high conjugation efficiency of Ang2 to the particle surface, and an encapsulation efficiency of more than 70% of CDDP and TMZ. In vitro models, including BBB hCMEC/D3 cell tight monolayer, 3D BBB cell spheroid, and microfluidic BBB/GBM-on-a-chip models with cocultured BBB and glioblastoma cells, were employed to study the efficiency of the developed cubosomes to cross the BBB and showed that Ang2-functionalized cubosomes can penetrate the BBB more effectively. Furthermore, Ang2-functionalized cubosomes showed significantly higher uptake by U87 glioblastoma cells, with a 3-fold increase observed in the BBB/GBM-on-a-chip model as compared to that of the bare cubosomes. Additionally, the in vivo biodistribution showed that Ang2 modification could significantly enhance the brain accumulation of cubosomes in comparison to that of non-functionalized particles. Moreover, CDDP-loaded Ang2-functionalized cubosomes presented an enhanced toxic effect on U87 spheroids. These findings suggest that the developed Ang2-cubosomes are prospective for improved BBB crossing and enhanced delivery of therapeutics to glioblastoma and are worth pursuing further as a potential application of nanomedicine for GBM treatment.
Collapse
Affiliation(s)
- Xudong Cai
- School of Science, STEM College, RMIT University, Melbourne 3000, VIC, Australia
| | - Ahmed Refaat
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, VIC, Australia
| | - Poh-Yi Gan
- Department of Medicine, Centre for Inflammatory Diseases, Monash University, 246 Clayton Rd, Clayton 3168, VIC, Australia
| | - Bo Fan
- School of Chemistry, Monash University, Clayton 3800, VIC, Australia
| | - Haitao Yu
- School of Science, STEM College, RMIT University, Melbourne 3000, VIC, Australia
| | - San H Thang
- School of Chemistry, Monash University, Clayton 3800, VIC, Australia
| | - Calum J Drummond
- School of Science, STEM College, RMIT University, Melbourne 3000, VIC, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne 3052, VIC, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton 3168, Victoria, Australia
- Department of Materials Science & Engineering, Monash University, Clayton 3168, Victoria, Australia
| | - Nhiem Tran
- School of Science, STEM College, RMIT University, Melbourne 3000, VIC, Australia
| | - Jiali Zhai
- School of Science, STEM College, RMIT University, Melbourne 3000, VIC, Australia
| |
Collapse
|
10
|
Lindemann MC, Glänzer L, Roeth AA, Schmitz-Rode T, Slabu I. Towards Realistic 3D Models of Tumor Vascular Networks. Cancers (Basel) 2023; 15:5352. [PMID: 38001612 PMCID: PMC10670125 DOI: 10.3390/cancers15225352] [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: 08/28/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
For reliable in silico or in vitro investigations in, for example, biosensing and drug delivery applications, accurate models of tumor vascular networks down to the capillary size are essential. Compared to images acquired with conventional medical imaging techniques, digitalized histological tumor slices have a higher resolution, enabling the delineation of capillaries. Volume rendering procedures can then be used to generate a 3D model. However, the preparation of such slices leads to misalignments in relative slice orientation between consecutive slices. Thus, image registration algorithms are necessary to re-align the slices. Here, we present an algorithm for the registration and reconstruction of a vascular network from histologic slices applied to 169 tumor slices. The registration includes two steps. First, consecutive images are incrementally pre-aligned using feature- and area-based transformations. Second, using the previous transformations, parallel registration for all images is enabled. Combining intensity- and color-based thresholds along with heuristic analysis, vascular structures are segmented. A 3D interpolation technique is used for volume rendering. This results in a 3D vascular network with approximately 400-450 vessels with diameters down to 25-30 µm. A delineation of vessel structures with close distance was limited in areas of high structural density. Improvement can be achieved by using images with higher resolution and or machine learning techniques.
Collapse
Affiliation(s)
- Max C. Lindemann
- Institute of Applied Medical Engineering, Helmholtz Institute, Medical Faculty, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany (L.G.); (T.S.-R.)
| | - Lukas Glänzer
- Institute of Applied Medical Engineering, Helmholtz Institute, Medical Faculty, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany (L.G.); (T.S.-R.)
| | - Anjali A. Roeth
- Department of General, Visceral and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany
- Department of Surgery, Maastricht University, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Thomas Schmitz-Rode
- Institute of Applied Medical Engineering, Helmholtz Institute, Medical Faculty, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany (L.G.); (T.S.-R.)
| | - Ioana Slabu
- Institute of Applied Medical Engineering, Helmholtz Institute, Medical Faculty, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany (L.G.); (T.S.-R.)
| |
Collapse
|
11
|
Hendricks JM, Doubravsky CE, Wehri E, Li Z, Roberts MA, Deol KK, Lange M, Lasheras-Otero I, Momper JD, Dixon SJ, Bersuker K, Schaletzky J, Olzmann JA. Identification of structurally diverse FSP1 inhibitors that sensitize cancer cells to ferroptosis. Cell Chem Biol 2023; 30:1090-1103.e7. [PMID: 37178691 PMCID: PMC10524360 DOI: 10.1016/j.chembiol.2023.04.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/07/2023] [Accepted: 04/12/2023] [Indexed: 05/15/2023]
Abstract
Ferroptosis is a regulated form of cell death associated with the iron-dependent accumulation of phospholipid hydroperoxides. Inducing ferroptosis is a promising approach to treat therapy-resistant cancer. Ferroptosis suppressor protein 1 (FSP1) promotes ferroptosis resistance in cancer by generating the antioxidant form of coenzyme Q10 (CoQ). Despite the important role of FSP1, few molecular tools exist that target the CoQ-FSP1 pathway. Through a series of chemical screens, we identify several structurally diverse FSP1 inhibitors. The most potent of these compounds, ferroptosis sensitizer 1 (FSEN1), is an uncompetitive inhibitor that acts selectively through on-target inhibition of FSP1 to sensitize cancer cells to ferroptosis. Furthermore, a synthetic lethality screen reveals that FSEN1 synergizes with endoperoxide-containing ferroptosis inducers, including dihydroartemisinin, to trigger ferroptosis. These results provide new tools that catalyze the exploration of FSP1 as a therapeutic target and highlight the value of combinatorial therapeutic regimes targeting FSP1 and additional ferroptosis defense pathways.
Collapse
Affiliation(s)
- Joseph M Hendricks
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Cody E Doubravsky
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Eddie Wehri
- The Henry Wheeler Center for Emerging and Neglected Diseases, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Zhipeng Li
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Melissa A Roberts
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kirandeep K Deol
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Mike Lange
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Irene Lasheras-Otero
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Jeremiah D Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Kirill Bersuker
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Julia Schaletzky
- The Henry Wheeler Center for Emerging and Neglected Diseases, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Miller Institute for Basic Research in Science, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub - San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
12
|
van Rijt A, Stefanek E, Valente K. Preclinical Testing Techniques: Paving the Way for New Oncology Screening Approaches. Cancers (Basel) 2023; 15:4466. [PMID: 37760435 PMCID: PMC10526899 DOI: 10.3390/cancers15184466] [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: 07/28/2023] [Revised: 08/24/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Prior to clinical trials, preclinical testing of oncology drug candidates is performed by evaluating drug candidates with in vitro and in vivo platforms. For in vivo testing, animal models are used to evaluate the toxicity and efficacy of drug candidates. However, animal models often display poor translational results as many drugs that pass preclinical testing fail when tested with humans, with oncology drugs exhibiting especially poor acceptance rates. The FDA Modernization Act 2.0 promotes alternative preclinical testing techniques, presenting the opportunity to use higher complexity in vitro models as an alternative to in vivo testing, including three-dimensional (3D) cell culture models. Three-dimensional tissue cultures address many of the shortcomings of 2D cultures by more closely replicating the tumour microenvironment through a combination of physiologically relevant drug diffusion, paracrine signalling, cellular phenotype, and vascularization that can better mimic native human tissue. This review will discuss the common forms of 3D cell culture, including cell spheroids, organoids, organs-on-a-chip, and 3D bioprinted tissues. Their advantages and limitations will be presented, aiming to discuss the use of these 3D models to accurately represent human tissue and as an alternative to animal testing. The use of 3D culture platforms for preclinical drug development is expected to accelerate as these platforms continue to improve in complexity, reliability, and translational predictivity.
Collapse
Affiliation(s)
- Antonia van Rijt
- Biomedical Engineering Program, University of Victoria, Victoria, BC V8P 5C2, Canada;
| | - Evan Stefanek
- VoxCell BioInnovation Inc., Victoria, BC V8T 5L2, Canada;
| | - Karolina Valente
- Biomedical Engineering Program, University of Victoria, Victoria, BC V8P 5C2, Canada;
| |
Collapse
|
13
|
Manikandan C, Jaiswal AK. Scaffold-based spheroid models of glioblastoma multiforme and its use in drug screening. Biotechnol Bioeng 2023. [PMID: 37366303 DOI: 10.1002/bit.28481] [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: 03/17/2023] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023]
Abstract
Among several types of brain cancers, glioblastoma multiforme (GBM) is a terminal and aggressive disease with a median survival of 15 months despite the most intensive surgery and chemotherapy. Preclinical models that accurately reproduce the tumor microenvironment are vital for developing new therapeutic alternatives. Understanding the complicated interactions between cells and their surroundings is essential to comprehend the tumor's microenvironment, however the monolayer cell culture approach falls short. Numerous approaches are used to develop GBM cells into tumor spheroids, while scaffold-based spheroids provides the opportunity to investigate the synergies between cells as well as cells and the matrix. This review summarizes the development of various scaffold-based GBM spheroid models and the prospective for their use as drug testing systems.
Collapse
Affiliation(s)
- Ceera Manikandan
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
- Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, India
| | - Amit Kumar Jaiswal
- Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
14
|
Al Hrout A, Levesque MP, Chahwan R. Investigating the tumor-immune microenvironment through extracellular vesicles from frozen patient biopsies and 3D cultures. Front Immunol 2023; 14:1176175. [PMID: 37304281 PMCID: PMC10248017 DOI: 10.3389/fimmu.2023.1176175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/04/2023] [Indexed: 06/13/2023] Open
Abstract
Melanomas are highly immunogenic tumors that have been shown to activate the immune response. Nonetheless, a significant portion of melanoma cases are either unresponsive to immunotherapy or relapsed due to acquired resistance. During melanomagenesis, melanoma and immune cells undergo immunomodulatory mechanisms that aid in immune resistance and evasion. The crosstalk within melanoma microenvironment is facilitated through the secretion of soluble factors, growth factors, cytokines, and chemokines. In addition, the release and uptake of secretory vesicles known as extracellular vesicles (EVs) play a key role in shaping the tumor microenvironment (TME). Melanoma-derived EVs have been implicated in immune suppression and escape, promoting tumor progression. In the context of cancer patients, EVs are usually isolated from biofluids such as serum, urine, and saliva. Nonetheless, this approach neglects the fact that biofluid-derived EVs reflect not only the tumor, but also include contributions from different organs and cell types. For that, isolating EVs from tissue samples allows for studying different cell populations resident at the tumor site, such as tumor-infiltrating lymphocytes and their secreted EVs, which play a central anti-tumor role. Herein, we outline the first instance of a method for EV isolation from frozen tissue samples at high purity and sensitivity that can be easily reproduced without the need for complicated isolation methods. Our method of processing the tissue not only circumvents the need for hard-to-acquire freshly isolated tissue samples, but also preserves EV surface proteins which allows for multiplex surface markers profiling. Tissue-derived EVs provide insight into the physiological role of EVs enrichment at tumor sites, which can be overlooked when studying circulating EVs coming from different sources. Tissue-derived EVs could be further characterized in terms of their genomics and proteomics to identify possible mechanisms for regulating the TME. Additionally, identified markers could be correlated to overall patient survival and disease progression for prognostic purposes.
Collapse
Affiliation(s)
- Ala’a Al Hrout
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Mitchell P. Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Richard Chahwan
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Biosensor integrated brain-on-a-chip platforms: Progress and prospects in clinical translation. Biosens Bioelectron 2023; 225:115100. [PMID: 36709589 DOI: 10.1016/j.bios.2023.115100] [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: 10/16/2022] [Revised: 01/07/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023]
Abstract
Because of the brain's complexity, developing effective treatments for neurological disorders is a formidable challenge. Research efforts to this end are advancing as in vitro systems have reached the point that they can imitate critical components of the brain's structure and function. Brain-on-a-chip (BoC) was first used for microfluidics-based systems with small synthetic tissues but has expanded recently to include in vitro simulation of the central nervous system (CNS). Defining the system's qualifying parameters may improve the BoC for the next generation of in vitro platforms. These parameters show how well a given platform solves the problems unique to in vitro CNS modeling (like recreating the brain's microenvironment and including essential parts like the blood-brain barrier (BBB)) and how much more value it offers than traditional cell culture systems. This review provides an overview of the practical concerns of creating and deploying BoC systems and elaborates on how these technologies might be used. Not only how advanced biosensing technologies could be integrated with BoC system but also how novel approaches will automate assays and improve point-of-care (PoC) diagnostics and accurate quantitative analyses are discussed. Key challenges providing opportunities for clinical translation of BoC in neurodegenerative disorders are also addressed.
Collapse
|
16
|
Ghalandary M, Gao Y, Amend D, Kutkaite G, Vick B, Spiekermann K, Rothenberg-Thurley M, Metzeler KH, Marcinek A, Subklewe M, Menden MP, Jurinovic V, Bahrami E, Jeremias I. WT1 and DNMT3A play essential roles in the growth of certain patient AML cells in mice. Blood 2023; 141:955-960. [PMID: 36256915 DOI: 10.1182/blood.2022016411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/16/2022] [Accepted: 09/25/2022] [Indexed: 11/20/2022] Open
Affiliation(s)
- Maryam Ghalandary
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Yuqiao Gao
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Diana Amend
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Ginte Kutkaite
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- Department of Biology, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Binje Vick
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- German Cancer Consortium, Partner Site Munich, Munich, Germany
| | - Karsten Spiekermann
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, Ludwig Maximilians University, Munich, Germany
| | - Maja Rothenberg-Thurley
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, Ludwig Maximilians University, Munich, Germany
| | - Klaus H Metzeler
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, Ludwig Maximilians University, Munich, Germany
- Department of Hematology and Cell Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Anetta Marcinek
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, Ludwig Maximilians University, Munich, Germany
| | - Marion Subklewe
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, Ludwig Maximilians University, Munich, Germany
| | - Michael P Menden
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- Department of Biology, Ludwig-Maximilians University Munich, Martinsried, Germany
- German Centre for Diabetes Research, Neuherberg, Germany
| | - Vindi Jurinovic
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Ehsan Bahrami
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Irmela Jeremias
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- German Cancer Consortium, Partner Site Munich, Munich, Germany
- Department of Pediatrics, University Hospital, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
17
|
Mierke CT. Physical and biological advances in endothelial cell-based engineered co-culture model systems. Semin Cell Dev Biol 2023; 147:58-69. [PMID: 36732105 DOI: 10.1016/j.semcdb.2023.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023]
Abstract
Scientific knowledge in the field of cell biology and mechanobiology heavily leans on cell-based in vitro experiments and models that favor the examination and comprehension of certain biological processes and occurrences across a variety of environments. Cell culture assays are an invaluable instrument for a vast spectrum of biomedical and biophysical investigations. The quality of experimental models in terms of simplicity, reproducibility, and combinability with other methods, and in particular the scale at which they depict cell fate in native tissues, is critical to advancing the knowledge of the comprehension of cell-cell and cell-matrix interactions in tissues and organs. Typically, in vitro models are centered on the experimental tinkering of mammalian cells, most often cultured as monolayers on planar, two-dimensional (2D) materials. Notwithstanding the significant advances and numerous findings that have been accomplished with flat biology models, their usefulness for generating further new biological understanding is constrained because the simple 2D setting does not reproduce the physiological response of cells in natural living tissues. In addition, the co-culture systems in a 2D stetting weakly mirror their natural environment of tissues and organs. Significant advances in 3D cell biology and matrix engineering have resulted in the creation and establishment of a new type of cell culture shapes that more accurately represents the in vivo microenvironment and allows cells and their interactions to be analyzed in a biomimetic approach. Contemporary biomedical and biophysical science has novel advances in technology that permit the design of more challenging and resilient in vitro models for tissue engineering, with a particular focus on scaffold- or hydrogel-based formats, organotypic cultures, and organs-on-chips, which cover the purposes of co-cultures. Even these complex systems must be kept as simplified as possible in order to grasp a particular section of physiology too very precisely. In particular, it is highly appreciated that they bridge the space between conventional animal research and human (patho)physiology. In this review, the recent progress in 3D biomimetic culturation is presented with a special focus on co-cultures, with an emphasis on the technological building blocks and endothelium-based co-culture models in cancer research that are available for the development of more physiologically relevant in vitro models of human tissues under normal and diseased conditions. Through applications and samples of various physiological and disease models, it is possible to identify the frontiers and future engagement issues that will have to be tackled to integrate synthetic biomimetic culture systems far more successfully into biomedical and biophysical investigations.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, Leipzig, Germany.
| |
Collapse
|
18
|
Evaluation of X-ray and carbon-ion beam irradiation with chemotherapy for the treatment of cervical adenocarcinoma cells in 2D and 3D cultures. Cancer Cell Int 2022; 22:391. [PMID: 36494817 PMCID: PMC9733259 DOI: 10.1186/s12935-022-02810-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cervical cancer is the second most common cancer in women and causes more than 250,000 deaths worldwide. Among these, the incidence of cervical adenocarcinomas is increasing. Cervical adenocarcinoma is not only difficult to detect and prevent in the early stages with screening, but it is also resistant to chemotherapy and radiotherapy, and its prognosis worsens significantly as the disease progresses. Furthermore, when recurrence or metastasis is observed, treatment options are limited and there is no curative treatment. Recently, heavy-particle radiotherapy has attracted attention owing to its high tumor control and minimal damage to normal tissues. In addition, heavy particle irradiation is effective for cancer stem cells and hypoxic regions, which are difficult to treat. METHODS In this study, we cultured cervical adenocarcinoma cell lines (HeLa and HCA-1) in two-dimensional (2D) or three-dimensional (3D) spheroid cultures and evaluated the effects of X-ray and carbon-ion (C-ion) beams. RESULTS X-ray irradiation decreased the cell viability in a dose-dependent manner in 2D cultures, whereas this effect was attenuated in 3D spheroid cultures. In contrast, C-ion irradiation demonstrated the same antitumor effect in 3D spheroid cultures as in 2D cultures. In 3D spheroid cultures, X-rays and anticancer drugs are attenuated because of hypoxia inside the spheroids. However, the impact of the C-ion beam was almost the same as that of the 2D culture, because heavy-particle irradiation was not affected by hypoxia. CONCLUSION These results suggest that heavy-particle radiotherapy may be a new therapeutic strategy for overcoming the resistance of cervical adenocarcinoma to treatment.
Collapse
|
19
|
Esposito R, Polidori T, Meise DF, Pulido-Quetglas C, Chouvardas P, Forster S, Schaerer P, Kobel A, Schlatter J, Kerkhof E, Roemmele M, Rice ES, Zhu L, Lanzós A, Guillen-Ramirez HA, Basile G, Carrozzo I, Vancura A, Ullrich S, Andrades A, Harvey D, Medina PP, Ma PC, Haefliger S, Wang X, Martinez I, Ochsenbein AF, Riether C, Johnson R. Multi-hallmark long noncoding RNA maps reveal non-small cell lung cancer vulnerabilities. CELL GENOMICS 2022; 2:100171. [PMID: 36778670 PMCID: PMC9903773 DOI: 10.1016/j.xgen.2022.100171] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 06/15/2022] [Accepted: 08/01/2022] [Indexed: 12/24/2022]
Abstract
Long noncoding RNAs (lncRNAs) are widely dysregulated in cancer, yet their functional roles in cancer hallmarks remain unclear. We employ pooled CRISPR deletion to perturb 831 lncRNAs detected in KRAS-mutant non-small cell lung cancer (NSCLC) and measure their contribution to proliferation, chemoresistance, and migration across two cell backgrounds. Integrative analysis of these data outperforms conventional "dropout" screens in identifying cancer genes while prioritizing disease-relevant lncRNAs with pleiotropic and background-independent roles. Altogether, 80 high-confidence oncogenic lncRNAs are active in NSCLC, which tend to be amplified and overexpressed in tumors. A follow-up antisense oligonucleotide (ASO) screen shortlisted two candidates, Cancer Hallmarks in Lung LncRNA 1 (CHiLL1) and GCAWKR, whose knockdown consistently suppressed cancer hallmarks in two- and three-dimension tumor models. Molecular phenotyping reveals that CHiLL1 and GCAWKR control cellular-level phenotypes via distinct transcriptional networks. This work reveals a multi-dimensional functional lncRNA landscape underlying NSCLC that contains potential therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Roberta Esposito
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso” CNR, Naples 80131, Italy
| | - Taisia Polidori
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern 3012, Switzerland
| | - Dominik F. Meise
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Carlos Pulido-Quetglas
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern 3012, Switzerland
| | - Panagiotis Chouvardas
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Stefan Forster
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Paulina Schaerer
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Andrea Kobel
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Juliette Schlatter
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Erik Kerkhof
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Michaela Roemmele
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Emily S. Rice
- Department of Microbiology, Immunology, and Cell Biology, Morgantown, WV, USA
| | - Lina Zhu
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
| | - Andrés Lanzós
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern 3012, Switzerland
| | - Hugo A. Guillen-Ramirez
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
- School of Biology and Environmental Science, University College Dublin, Dublin D04 V1W8, Ireland
- Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin D04 V1W8, Ireland
| | - Giulia Basile
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Irene Carrozzo
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Adrienne Vancura
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Sebastian Ullrich
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology (BIST), Barcelona, Catalonia 08003, Spain
| | - Alvaro Andrades
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada 18016, Spain
- Instituto de Investigación Biosanitaria, Granada 18014, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Granada 18071, Spain
| | - Dylan Harvey
- School of Biology and Environmental Science, University College Dublin, Dublin D04 V1W8, Ireland
| | - Pedro P. Medina
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada 18016, Spain
- Instituto de Investigación Biosanitaria, Granada 18014, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Granada 18071, Spain
| | | | - Simon Haefliger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Xin Wang
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Ivan Martinez
- Department of Microbiology, Immunology, and Cell Biology, Morgantown, WV, USA
| | - Adrian F. Ochsenbein
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Carsten Riether
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
| | - Rory Johnson
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3010 Switzerland
- Department for BioMedical Research, University of Bern, Bern 3008, Switzerland
- School of Biology and Environmental Science, University College Dublin, Dublin D04 V1W8, Ireland
- Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin D04 V1W8, Ireland
| |
Collapse
|
20
|
Li Z, Ferguson L, Deol KK, Roberts MA, Magtanong L, Hendricks JM, Mousa GA, Kilinc S, Schaefer K, Wells JA, Bassik MC, Goga A, Dixon SJ, Ingolia NT, Olzmann JA. Ribosome stalling during selenoprotein translation exposes a ferroptosis vulnerability. Nat Chem Biol 2022; 18:751-761. [PMID: 35637349 PMCID: PMC9469796 DOI: 10.1038/s41589-022-01033-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/06/2022] [Indexed: 01/23/2023]
Abstract
The selenoprotein glutathione peroxidase 4 (GPX4) prevents ferroptosis by converting lipid peroxides into nontoxic lipid alcohols. GPX4 has emerged as a promising therapeutic target for cancer treatment, but some cancer cells are resistant to ferroptosis triggered by GPX4 inhibition. Using a chemical-genetic screen, we identify LRP8 (also known as ApoER2) as a ferroptosis resistance factor that is upregulated in cancer. Loss of LRP8 decreases cellular selenium levels and the expression of a subset of selenoproteins. Counter to the canonical hierarchical selenoprotein regulatory program, GPX4 levels are strongly reduced due to impaired translation. Mechanistically, low selenium levels result in ribosome stalling at the inefficiently decoded GPX4 selenocysteine UGA codon, leading to ribosome collisions, early translation termination and proteasomal clearance of the N-terminal GPX4 fragment. These findings reveal rewiring of the selenoprotein hierarchy in cancer cells and identify ribosome stalling and collisions during GPX4 translation as ferroptosis vulnerabilities in cancer.
Collapse
Affiliation(s)
- Zhipeng Li
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Lucas Ferguson
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Kirandeep K Deol
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Melissa A Roberts
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | | | - Joseph M Hendricks
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Gergey Alzaem Mousa
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Seda Kilinc
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Kaitlin Schaefer
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Andrei Goga
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Nicholas T Ingolia
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
- Miller Institute for Basic Research in Science, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
21
|
Al Hrout A, Cervantes-Gracia K, Chahwan R, Amin A. Modelling liver cancer microenvironment using a novel 3D culture system. Sci Rep 2022; 12:8003. [PMID: 35568708 PMCID: PMC9107483 DOI: 10.1038/s41598-022-11641-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 04/27/2022] [Indexed: 12/22/2022] Open
Abstract
The tumor microenvironment and its contribution to tumorigenesis has been a focal highlight in recent years. A two-way communication between the tumor and the surrounding microenvironment sustains and contributes to the growth and metastasis of tumors. Progression and metastasis of hepatocellular carcinoma (HCC) have been reported to be exceedingly influenced by diverse microenvironmental cues. In this study, we present a 3D-culture model of liver cancer to better mimic in vivo tumor settings. By creating novel 3D co-culture model that combines free-floating and scaffold-based 3D-culture techniques of liver cancer cells and fibroblasts, we aimed to establish a simple albeit reproducible ex vivo cancer microenvironment model that captures tumor-stroma interactions. The model presented herein exhibited unique gene expression and protein expression profiles when compared to 2D and 3D mono-cultures of liver cancer cells. Our results showed that in vivo like conditions cannot be mimicked by simply growing cancer cells as spheroids, but by co-culturing them with 3D fibroblast with which they were able to crosstalk. This was evident by the upregulation of several pathways involved in HCC, and the increase in secreted factors by co-cultured cancer cells, many of which are also involved in tumor-stroma interactions. Compared to the conventional 2D culture, the proposed model exhibits an increase in the expression of genes associated with development, progression, and poor prognosis of HCC. Our results correlated with an aggressive outcome that better mirrors in vivo HCC, and therefore, a more reliable platform for molecular understanding of HCC.
Collapse
Affiliation(s)
- Ala'a Al Hrout
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Biology Department, College of Science, UAE University, P.O. Box 15551, Al-Ain, United Arab Emirates
| | - Karla Cervantes-Gracia
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Richard Chahwan
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| | - Amr Amin
- Biology Department, College of Science, UAE University, P.O. Box 15551, Al-Ain, United Arab Emirates.
- The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
22
|
Salerno S, Ståhlberg A, Holdfeldt A, Bexe Lindskog E, Landberg G. 5-fluorouracil treatment of patient-derived scaffolds from colorectal cancer reveal clinically critical information. J Transl Med 2022; 20:209. [PMID: 35562738 PMCID: PMC9102939 DOI: 10.1186/s12967-022-03423-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/02/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Colorectal cancer is a commonly diagnosed cancer worldwide. Unfortunately, many patients do not respond to standard chemotherapy treatments and develop disease relapse and metastases. Besides cancer cell specific genetic changes, heterogeneity in the tumor microenvironment contribute to the clinical presentation of the disease and can potentially also influence drug resistance. By using a recently developed patient-derived scaffold method monitoring how a standardized reporter cancer cell line adapts to various microenvironments treated with chemotherapy, we wanted to clarify how individual patient specific microenvironments influence the chemotherapy response in colorectal cancer. METHODS Surgically resected colorectal cancer specimens from 89 patients were decellularized to produce patient-derived scaffold, which were seeded with HT29 cells, cultured for 3 weeks, and treated with 5-fluorouracil. Gene expression changes of adapted and treated HT29 cells were monitored by qPCR and compared with clinical parameters including disease-free survival. RESULTS The effects of 5-fluorouracil treatment varied between different patient-derived scaffold, but generally induced a reduced expression of proliferation genes and increased expression of pluripotency and epithelial-to-mesenchymal transition genes. Interestingly, patient-derived scaffold cultures obtained from patients with disease recurrences showed a significantly less pronounced anti-proliferative effect of 5-fluorouracil and more pronounced increase of pluripotency, with MKI67 and POU5F1 being among the most significant genes linked to disease relapse in colorectal cancer. CONCLUSIONS Colorectal patient-derived scaffold can decode clinically relevant tumor microenvironmental influence of 5-fluorouracil treatment effects opening up for optimized precision medicine in colorectal cancer treatment.
Collapse
Affiliation(s)
- Simona Salerno
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anders Ståhlberg
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Genetics and Genomics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - André Holdfeldt
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Elinor Bexe Lindskog
- Department of Surgery, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.,Surgical Oncology Laboratory, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Göran Landberg
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden. .,Department of Pathology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
23
|
Liu RJ, Xu ZP, Li SY, Yu JJ, Feng NH, Xu B, Chen M. BAP1-Related ceRNA (NEAT1/miR-10a-5p/SERPINE1) Promotes Proliferation and Migration of Kidney Cancer Cells. Front Oncol 2022; 12:852515. [PMID: 35425712 PMCID: PMC9004599 DOI: 10.3389/fonc.2022.852515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/25/2022] [Indexed: 01/09/2023] Open
Abstract
Background BAP1 is an important tumor suppressor involved in various biological processes and is commonly lost or inactivated in clear-cell renal cell carcinoma (ccRCC). However, the role of the BAP1-deficient tumor competing endogenous RNA (ceRNA) network involved in ccRCC remains unclear. Thus, this study aims to investigate the prognostic BAP1-related ceRNA in ccRCC. Methods Raw data was obtained from the TCGA and the differentially expressed genes were screened to establish a BAP1-related ceRNA network. Subsequently, the role of the ceRNA axis was validated using phenotypic experiments. Dual-luciferase reporter assays and fluorescence in situ hybridization (FISH) assays were used to confirm the ceRNA network. Results Nuclear enriched abundant transcript 1 (NEAT1) expression was significantly increased in kidney cancer cell lines. NEAT1 knockdown significantly inhibited cell proliferation and migration, which could be reversed by miR-10a-5p inhibitor. Dual-luciferase reporter assay confirmed miR-10a-5p as a common target of NEAT1 and Serine protease inhibitor family E member 1 (SERPINE1). FISH assays revealed the co-localization of NEAT1 and miR-10a-5p in the cytoplasm. Additionally, the methylation level of SERPINE1 in ccRCC was significantly lower than that in normal tissues. Furthermore, SERPINE1 expression was positively correlated with multiple immune cell infiltration levels. Conclusions In BAP1-deficient ccRCC, NEAT1 competitively binds to miR-10a-5p, indirectly upregulating SERPINE1 expression to promote kidney cancer cell proliferation. Furthermore, NEAT1/miR-10a-5p/SERPINE1 were found to be independent prognostic factors of ccRCC.
Collapse
Affiliation(s)
- Rui-Ji Liu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China
| | - Zhi-Peng Xu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China
| | - Shu-Ying Li
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital affiliate to School of Medicine, UESTC, Chengdu, China
| | - Jun-Jie Yu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China
| | - Ning-Han Feng
- Department of Urology, Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, China
| | - Bin Xu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China
| | - Ming Chen
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.,Nanjing Lishui District People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| |
Collapse
|
24
|
Extracellular ATP promotes breast cancer chemoresistance via HIF-1α signaling. Cell Death Dis 2022; 13:199. [PMID: 35236823 PMCID: PMC8891368 DOI: 10.1038/s41419-022-04647-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/29/2022] [Accepted: 02/11/2022] [Indexed: 12/27/2022]
Abstract
We have previously demonstrated that extracellular adenosine 5'-triphosphate (ATP) promotes breast cancer cell chemoresistance. However, the underlying mechanism remains unclear. Using a cDNA microarray, we demonstrated that extracellular ATP can stimulate hypoxia-inducible factor (HIF) signaling. In this study, we report that hypoxia-inducible factor 1α (HIF-1α) was upregulated after ATP treatment and mediated the ATP-driven chemoresistance process. We aimed to investigate the mechanisms and identify potential clinically relevant targets that are involved. Using mass spectrometry, we found that aldolase A (ALDOA) interacts with HIF-1α and increases HIF-1α expression. We then demonstrated that STAT3-ALDOA mediates ATP-HIF-1α signaling and upregulates the HIF-1 target genes adrenomedullin (ADM) and phosphoinositide-dependent kinase-1 (PDK1). Moreover, we show that PI3K/AKT acts upstream of HIF-1α in ATP signaling and contributes to chemoresistance in breast cancer cells. In addition, HIF-1α-knockdown or treatment with direct HIF inhibitors combined with the ATP hydrolase apyrase in MDA-MB-231 cells induced enhanced drug sensitivity in nude BALB/c mice. We then used in vitro spheroid formation assays to demonstrate the significance of ATP-HIF-1α in mediating chemoresistance. Furthermore, considering that indirect HIF inhibitors are effective in clinical cancer therapy, we treated tumor-bearing BALB/c mice with STAT3 and PI3K/AKT inhibitors and found that the dual-targeting strategy sensitized breast cancer to cisplatin. Finally, using breast cancer tissue microarrays, we found that ATP-HIF-1α signaling is associated with cancer progression, poor prognosis, and resistance to chemotherapy. Taken together, we suggest that HIF-1α signaling is vital in ATP-driven chemoresistance and may serve as a potential target for breast cancer therapies.
Collapse
|
25
|
Campioni G, Pasquale V, Busti S, Ducci G, Sacco E, Vanoni M. An Optimized Workflow for the Analysis of Metabolic Fluxes in Cancer Spheroids Using Seahorse Technology. Cells 2022; 11:cells11050866. [PMID: 35269488 PMCID: PMC8909358 DOI: 10.3390/cells11050866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Three-dimensional cancer models, such as spheroids, are increasingly being used to study cancer metabolism because they can better recapitulate the molecular and physiological aspects of the tumor architecture than conventional monolayer cultures. Although Agilent Seahorse XFe96 (Agilent Technologies, Santa Clara, CA, United States) is a valuable technology for studying metabolic alterations occurring in cancer cells, its application to three-dimensional cultures is still poorly optimized. We present a reliable and reproducible workflow for the Seahorse metabolic analysis of three-dimensional cultures. An optimized protocol enables the formation of spheroids highly regular in shape and homogenous in size, reducing variability in metabolic parameters among the experimental replicates, both under basal and drug treatment conditions. High-resolution imaging allows the calculation of the number of viable cells in each spheroid, the normalization of metabolic parameters on a per-cell basis, and grouping of the spheroids as a function of their size. Multivariate statistical tests on metabolic parameters determined by the Mito Stress test on two breast cancer cell lines show that metabolic differences among the studied spheroids are mostly related to the cell line rather than to the size of the spheroid. The optimized workflow allows high-resolution metabolic characterization of three-dimensional cultures, their comparison with monolayer cultures, and may aid in the design and interpretation of (multi)drug protocols.
Collapse
Affiliation(s)
- Gloria Campioni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (V.P.); (S.B.); (G.D.); (E.S.)
- SYSBIO (Centre of Systems Biology), ISBE (Infrastructure Systems Biology Europe), 20126 Milan, Italy
| | - Valentina Pasquale
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (V.P.); (S.B.); (G.D.); (E.S.)
- SYSBIO (Centre of Systems Biology), ISBE (Infrastructure Systems Biology Europe), 20126 Milan, Italy
| | - Stefano Busti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (V.P.); (S.B.); (G.D.); (E.S.)
- SYSBIO (Centre of Systems Biology), ISBE (Infrastructure Systems Biology Europe), 20126 Milan, Italy
| | - Giacomo Ducci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (V.P.); (S.B.); (G.D.); (E.S.)
- SYSBIO (Centre of Systems Biology), ISBE (Infrastructure Systems Biology Europe), 20126 Milan, Italy
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (V.P.); (S.B.); (G.D.); (E.S.)
- SYSBIO (Centre of Systems Biology), ISBE (Infrastructure Systems Biology Europe), 20126 Milan, Italy
| | - Marco Vanoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (V.P.); (S.B.); (G.D.); (E.S.)
- SYSBIO (Centre of Systems Biology), ISBE (Infrastructure Systems Biology Europe), 20126 Milan, Italy
- Correspondence: ; Tel.: +39-02-6448-3525
| |
Collapse
|
26
|
Li J, Chen J, Bai H, Wang H, Hao S, Ding Y, Peng B, Zhang J, Li L, Huang W. An Overview of Organs-on-Chips Based on Deep Learning. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9869518. [PMID: 35136860 PMCID: PMC8795883 DOI: 10.34133/2022/9869518] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/08/2021] [Indexed: 12/15/2022]
Abstract
Microfluidic-based organs-on-chips (OoCs) are a rapidly developing technology in biomedical and chemical research and have emerged as one of the most advanced and promising in vitro models. The miniaturization, stimulated tissue mechanical forces, and microenvironment of OoCs offer unique properties for biomedical applications. However, the large amount of data generated by the high parallelization of OoC systems has grown far beyond the scope of manual analysis by researchers with biomedical backgrounds. Deep learning, an emerging area of research in the field of machine learning, can automatically mine the inherent characteristics and laws of "big data" and has achieved remarkable applications in computer vision, speech recognition, and natural language processing. The integration of deep learning in OoCs is an emerging field that holds enormous potential for drug development, disease modeling, and personalized medicine. This review briefly describes the basic concepts and mechanisms of microfluidics and deep learning and summarizes their successful integration. We then analyze the combination of OoCs and deep learning for image digitization, data analysis, and automation. Finally, the problems faced in current applications are discussed, and future perspectives and suggestions are provided to further strengthen this integration.
Collapse
Affiliation(s)
- Jintao Li
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Jie Chen
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, School of Electronics and Information Engineering, Anhui University, Hefei 230601, China
- 38th Research Institute of China Electronics Technology Group Corporation, Hefei 230088, China
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Haiwei Wang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Shiping Hao
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yang Ding
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Jing Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Lin Li
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech), Nanjing 211800, China
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech), Nanjing 211800, China
| |
Collapse
|
27
|
Tran TA, Kappelhoff J, Jüstel T, Anderson RR, Purschke M. UV emitting nanoparticles enhance the effect of ionizing radiation in 3D lung cancer spheroids. Int J Radiat Biol 2022; 98:1484-1494. [PMID: 35020574 DOI: 10.1080/09553002.2022.2027541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE Radiation therapy for cancer is limited by damage to surrounding normal tissues, and failure to completely eradicate a tumor. This study investigated a novel radiosensitizer, composed of lutetium phosphate nanoparticles doped with 1% praseodymium and 1.5% neodymium cations (LuPO4:Pr3+,Nd3+). During X-ray exposure, the particles emit UVC photons (200-280 nm), resulting in increased tumor cell death, by oxygen-independent UVC-induced damage. METHODS AND MATERIALS Specially designed LuPO4:Pr3+,Nd3+ nanoscintillator particles were characterized by dynamic light scattering, TEM and emission spectroscopy upon excitation. Cell death was determined by reduction in tumor spheroid growth over a 3-week period using a 3D A549 lung cancer model. Cell cycle was evaluated by flow cytometry and cell death pathways were assessed by Annexin V/PI stain as well as quantify apoptotic bodies. RESULTS Lung cancer cells expressed no long-term or non-specific toxicity when incubated with LuPO4:Pr3+,Nd3+ nanoscintillators. In contrast, there was significant growth inhibition of cell spheres treated with 2.5 mg/ml LuPO4:Pr3+,Nd3+ in combination with ionizing radiation (4 or 8 Gy X-ray), compared to radiation alone. A homogeneous distribution of small NPs throughout the entire sphere resulted in more pronounced lethality and growth inhibition, compared to particle distribution limited to the outer cell layers. Growth inhibition after the combined treatment was caused by necrosis, apoptosis and G2/M cell cycle arrest. CONCLUSIONS Newly designed UVC-emitting nanoscintillators (LuPO4:Pr3+,Nd3+) in combination with ionizing radiation cause tumor sphere growth inhibition by inducing cell cycle arrest, apoptosis and necrosis. UVC-emitting nanoparticles offer a promising new strategy for enhancing local tumor response to ionizing radiation treatment.
Collapse
Affiliation(s)
- Thao Anh Tran
- Wellman Center for Photomedicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA, United States.,Dept. Medicine, University of Geneva, Geneva, Switzerland
| | - Jan Kappelhoff
- Dept. Chemical Engineering, Münster University of Applied Sciences, Münster, Germany
| | - Thomas Jüstel
- Dept. Chemical Engineering, Münster University of Applied Sciences, Münster, Germany
| | - R Rox Anderson
- Wellman Center for Photomedicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA, United States
| | - Martin Purschke
- Wellman Center for Photomedicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA, United States.,Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, CA, USA
| |
Collapse
|
28
|
Barbosa MAG, Xavier CPR, Pereira RF, Petrikaitė V, Vasconcelos MH. 3D Cell Culture Models as Recapitulators of the Tumor Microenvironment for the Screening of Anti-Cancer Drugs. Cancers (Basel) 2021; 14:190. [PMID: 35008353 PMCID: PMC8749977 DOI: 10.3390/cancers14010190] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Today, innovative three-dimensional (3D) cell culture models have been proposed as viable and biomimetic alternatives for initial drug screening, allowing the improvement of the efficiency of drug development. These models are gaining popularity, given their ability to reproduce key aspects of the tumor microenvironment, concerning the 3D tumor architecture as well as the interactions of tumor cells with the extracellular matrix and surrounding non-tumor cells. The development of accurate 3D models may become beneficial to decrease the use of laboratory animals in scientific research, in accordance with the European Union's regulation on the 3R rule (Replacement, Reduction, Refinement). This review focuses on the impact of 3D cell culture models on cancer research, discussing their advantages, limitations, and compatibility with high-throughput screenings and automated systems. An insight is also given on the adequacy of the available readouts for the interpretation of the data obtained from the 3D cell culture models. Importantly, we also emphasize the need for the incorporation of additional and complementary microenvironment elements on the design of 3D cell culture models, towards improved predictive value of drug efficacy.
Collapse
Affiliation(s)
- Mélanie A. G. Barbosa
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Cristina P. R. Xavier
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Rúben F. Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Biofabrication Group, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickevičiaus g 9, LT-44307 Kaunas, Lithuania;
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania
| | - M. Helena Vasconcelos
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
29
|
|
30
|
Xie H, Appelt JW, Jenkins RW. Going with the Flow: Modeling the Tumor Microenvironment Using Microfluidic Technology. Cancers (Basel) 2021; 13:cancers13236052. [PMID: 34885161 PMCID: PMC8656483 DOI: 10.3390/cancers13236052] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/20/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The clinical success of cancer immunotherapy targeting immune checkpoints (e.g., PD-1, CTLA-4) has ushered in a new era of cancer therapeutics aimed at promoting antitumor immunity in hopes of offering durable clinical responses for patients with advanced, metastatic cancer. This success has also reinvigorated interest in developing tumor model systems that recapitulate key features of antitumor immune responses to complement existing in vivo tumor models. Patient-derived tumor models have emerged in recent years to facilitate study of tumor–immune dynamics. Microfluidic technology has enabled development of microphysiologic systems (MPSs) for the evaluation of the tumor microenvironment, which have shown early promise in studying tumor–immune dynamics. Further development of microfluidic-based “tumor-on-a-chip” MPSs to study tumor–immune interactions may overcome several key challenges currently facing tumor immunology. Abstract Recent advances in cancer immunotherapy have led a paradigm shift in the treatment of multiple malignancies with renewed focus on the host immune system and tumor–immune dynamics. However, intrinsic and acquired resistance to immunotherapy limits patient benefits and wider application. Investigations into the mechanisms of response and resistance to immunotherapy have demonstrated key tumor-intrinsic and tumor-extrinsic factors. Studying complex interactions with multiple cell types is necessary to understand the mechanisms of response and resistance to cancer therapies. The lack of model systems that faithfully recapitulate key features of the tumor microenvironment (TME) remains a challenge for cancer researchers. Here, we review recent advances in TME models focusing on the use of microfluidic technology to study and model the TME, including the application of microfluidic technologies to study tumor–immune dynamics and response to cancer therapeutics. We also discuss the limitations of current systems and suggest future directions to utilize this technology to its highest potential.
Collapse
Affiliation(s)
- Hongyan Xie
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (H.X.); (J.W.A.)
| | - Jackson W. Appelt
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (H.X.); (J.W.A.)
| | - Russell W. Jenkins
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (H.X.); (J.W.A.)
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Correspondence: ; Tel.: +617-726-9372; Fax: +844-542-5959
| |
Collapse
|
31
|
Becker L, Janssen N, Layland SL, Mürdter TE, Nies AT, Schenke-Layland K, Marzi J. Raman Imaging and Fluorescence Lifetime Imaging Microscopy for Diagnosis of Cancer State and Metabolic Monitoring. Cancers (Basel) 2021; 13:cancers13225682. [PMID: 34830837 PMCID: PMC8616063 DOI: 10.3390/cancers13225682] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Hurdles for effective tumor therapy are delayed detection and limited effectiveness of systemic drug therapies by patient-specific multidrug resistance. Non-invasive bioimaging tools such as fluorescence lifetime imaging microscopy (FLIM) and Raman-microspectroscopy have evolved over the last decade, providing the potential to be translated into clinics for early-stage disease detection, in vitro drug screening, and drug efficacy studies in personalized medicine. Accessing tissue- and cell-specific spectral signatures, Raman microspectroscopy has emerged as a diagnostic tool to identify precancerous lesions, cancer stages, or cell malignancy. In vivo Raman measurements have been enabled by recent technological advances in Raman endoscopy and signal-enhancing setups such as coherent anti-stokes Raman spectroscopy or surface-enhanced Raman spectroscopy. FLIM enables in situ investigations of metabolic processes such as glycolysis, oxidative stress, or mitochondrial activity by using the autofluorescence of co-enzymes NADH and FAD, which are associated with intrinsic proteins as a direct measure of tumor metabolism, cell death stages and drug efficacy. The combination of non-invasive and molecular-sensitive in situ techniques and advanced 3D tumor models such as patient-derived organoids or microtumors allows the recapitulation of tumor physiology and metabolism in vitro and facilitates the screening for patient-individualized drug treatment options.
Collapse
Affiliation(s)
- Lucas Becker
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
| | - Nicole Janssen
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Shannon L Layland
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, University of Tübingen, 72076 Tübingen, Germany
| | - Thomas E Mürdter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Anne T Nies
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Katja Schenke-Layland
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
- Cardiovascular Research Laboratories, Department of Medicine/Cardiology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90073, USA
| | - Julia Marzi
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
| |
Collapse
|
32
|
Calcium-Based Biomineralization: A Smart Approach for the Design of Novel Multifunctional Hybrid Materials. JOURNAL OF COMPOSITES SCIENCE 2021. [DOI: 10.3390/jcs5100278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Biomineralization consists of a complex cascade of phenomena generating hybrid nano-structured materials based on organic (e.g., polymer) and inorganic (e.g., hydroxyapatite) components. Biomineralization is a biomimetic process useful to produce highly biomimetic and biocompatible materials resembling natural hard tissues such as bones and teeth. In detail, biomimetic materials, composed of hydroxyapatite nanoparticles (HA) nucleated on an organic matrix, show extremely versatile chemical compositions and physical properties, which can be controlled to address specific challenges. Indeed, different parameters, including (i) the partial substitution of mimetic doping ions within the HA lattice, (ii) the use of different organic matrices, and (iii) the choice of cross-linking processes, can be finely tuned. In the present review, we mainly focused on calcium biomineralization. Besides regenerative medicine, these multifunctional materials have been largely exploited for other applications including 3D printable materials and in vitro three-dimensional (3D) models for cancer studies and for drug testing. Additionally, biomineralized multifunctional nano-particles can be involved in applications ranging from nanomedicine as fully bioresorbable drug delivery systems to the development of innovative and eco-sustainable UV physical filters for skin protection from solar radiations.
Collapse
|
33
|
Neufeld L, Yeini E, Reisman N, Shtilerman Y, Ben-Shushan D, Pozzi S, Madi A, Tiram G, Eldar-Boock A, Ferber S, Grossman R, Ram Z, Satchi-Fainaro R. Microengineered perfusable 3D-bioprinted glioblastoma model for in vivo mimicry of tumor microenvironment. SCIENCE ADVANCES 2021; 7:eabi9119. [PMID: 34407932 PMCID: PMC8373143 DOI: 10.1126/sciadv.abi9119] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/28/2021] [Indexed: 05/04/2023]
Abstract
Many drugs show promising results in laboratory research but eventually fail clinical trials. We hypothesize that one main reason for this translational gap is that current cancer models are inadequate. Most models lack the tumor-stroma interactions, which are essential for proper representation of cancer complexed biology. Therefore, we recapitulated the tumor heterogenic microenvironment by creating fibrin glioblastoma bioink consisting of patient-derived glioblastoma cells, astrocytes, and microglia. In addition, perfusable blood vessels were created using a sacrificial bioink coated with brain pericytes and endothelial cells. We observed similar growth curves, drug response, and genetic signature of glioblastoma cells grown in our 3D-bioink platform and in orthotopic cancer mouse models as opposed to 2D culture on rigid plastic plates. Our 3D-bioprinted model could be the basis for potentially replacing cell cultures and animal models as a powerful platform for rapid, reproducible, and robust target discovery; personalized therapy screening; and drug development.
Collapse
Affiliation(s)
- Lena Neufeld
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Eilam Yeini
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Noa Reisman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yael Shtilerman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dikla Ben-Shushan
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Asaf Madi
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Galia Tiram
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anat Eldar-Boock
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shiran Ferber
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Rachel Grossman
- Department of Neurosurgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Zvi Ram
- Department of Neurosurgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
- Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
34
|
Franchi-Mendes T, Lopes N, Brito C. Heterotypic Tumor Spheroids in Agitation-Based Cultures: A Scaffold-Free Cell Model That Sustains Long-Term Survival of Endothelial Cells. Front Bioeng Biotechnol 2021; 9:649949. [PMID: 34178955 PMCID: PMC8219978 DOI: 10.3389/fbioe.2021.649949] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/06/2021] [Indexed: 12/25/2022] Open
Abstract
Endothelial cells (ECs) are an important component of the tumor microenvironment, playing key roles in tumor development and progression that span from angiogenesis to immune regulation and drug resistance. Heterotypic tumor spheroids are one of the most widely used in vitro tumor microenvironment models, presenting improved recapitulation of tumor microenvironments compared to 2D cultures, in a simple and low-cost setup. Heterotypic tumor spheroid models incorporating endothelial cells have been proposed but present multiple limitations, such as the short culture duration typically obtained, the use of animal-derived matrices, and poor reproducibility; the diversity of culture conditions employed hinders comparison between studies and standardization of relevant culture parameters. Herein, we developed long-term cultures of triple heterotypic spheroids composed of the HCC1954 tumor cell line, human fibroblasts, and ECs. We explored culture parameters potentially relevant for EC maintenance, such as tumor cell line, seeding cell number, cell ratio, and agitation vs. static culture. In HCC1954-based spheroids, we observed maintenance of viable EC for up to 1 month of culture in agitation, with retention of the identity markers CD31 and von Willebrand factor. At the optimized tumor cell:fibroblast:EC ratio of 1:3:10, HCC1954-based spheroids had a higher EC area/total spheroid area at 1 month of culture than the other cell ratios tested. EC maintenance was tumor cell line-dependent, and in HCC1954-based spheroids it was also dependent on the presence of fibroblasts and agitation. Moreover, vascular endothelial growth factor (VEGF) supplementation was not required for maintenance of EC, as the factor was endogenously produced. ECs co-localized with fibroblasts, which accumulated preferentially in the core of the spheroids and secreted EC-relevant extracellular matrix proteins, such as collagen I and IV. This simple model setup does not rely on artificial or animal-derived scaffolds and can serve as a useful tool to explore the culture parameters influencing heterotypic spheroids, contributing to model standardization, as well as to explore molecular cross talk of ECs within the tumor microenvironment, and potentially its effects on drug response.
Collapse
Affiliation(s)
- Teresa Franchi-Mendes
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Nuno Lopes
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Oeiras, Portugal
| |
Collapse
|
35
|
Micalet A, Moeendarbary E, Cheema U. 3D In Vitro Models for Investigating the Role of Stiffness in Cancer Invasion. ACS Biomater Sci Eng 2021. [PMID: 34081437 DOI: 10.1021/acsbiomaterials.0c01530] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Tumorigenesis is attributed to the interactions of cancer cells with the tumor microenvironment through both biochemical cues and physical stimuli. Increased matrix deposition and realignment of the collagen fibers are detected by cancer cells, inducing epithelial-to-mesenchymal transition, which in turn stimulates cell motility and invasiveness. METHODS This review provides an overview of current research on the role of the physical microenvironment in cancer invasion. This was achieved by using a systematic approach and providing meta-analyses. Particular focus was placed on in vitro three-dimensional models of epithelial cancers. We investigated questions such as the effect of matrix stiffening, activation of stromal cells, and identified potential advances in mechano-based therapies. RESULTS Meta-analysis revealed that 64% of studies report cancer invasion promotion as stiffness increases, while 36% report the opposite. Experimental approaches and data interpretations were varied, each affecting the invasion of cancer differently. Examples are the experimental timeframes used (24 h to 21 days), the type of polymer used (24 types), and choice of cell line (33 cell lines). The stiffness of the 3D matrices varied from 0.5 to 300 kPa and 19% of these matrices' stiffness were outside commonly accepted physiological range. 100% of the studies outside biological stiffness range (above 20 kPa) report that stiffness does not promote cancer invasion. CONCLUSIONS Taking this analysis into account, we inform on the type of experimental approaches that could be the most relevant and provide what would be a standardized protocol and reporting strategy.
Collapse
Affiliation(s)
- Auxtine Micalet
- Department of Mechanical Engineering, University College London (UCL), Torrington Place, London, U.K. WC1E 6BT.,Division of Surgery and Interventional Sciences, UCL Centre for 3D Models of Health and Disease, University College London (UCL), Charles Bell House, London, U.K. W1W 7TS
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London (UCL), Torrington Place, London, U.K. WC1E 6BT.,Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, United States
| | - Umber Cheema
- Division of Surgery and Interventional Sciences, UCL Centre for 3D Models of Health and Disease, University College London (UCL), Charles Bell House, London, U.K. W1W 7TS
| |
Collapse
|
36
|
Almela T, Tayebi L, Moharamzadeh K. 3D Bioprinting for In Vitro Models of Oral Cancer: Toward Development and Validation. ACTA ACUST UNITED AC 2021; 22. [PMID: 34368488 DOI: 10.1016/j.bprint.2021.e00132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The tumor microenvironment (TME) of oral carcinomas has highly complex contents and a dynamic nature which is difficult to study using oversimplified two-dimensional (2D) cell culture systems. By contrast, three dimensional (3D) in vitro models such as spheroids, organoids, and scaffold-based constructs have been able to replicate tumors three-dimensionality and have allowed a better understanding of the role of various microenvironmental cues in the initiation and progression of cancer. However, the heterogeneity of TME cannot be fully reproduced by these traditional tissue engineering strategies since they are unable to control the organization of multiple cell types in a complex architecture. 3D bioprinting is an emerging field that can be leveraged to produce biomimetic and complex tissue structures. Bioprinting allows for controllable and precise placement of multicomponent bioinks composed of multiple biomaterials, different types of cells, and soluble factors according to the natural compartments of the target tissue, aiming to reproduce the equivalent of the complex tissue. As such, 3D bioprinting provides a unique opportunity to fabricate in vitro tumor models with a complexity similar to that of the in vivo oral carcinoma. This will facilitate a thorough investigation of cellular physiology, cancer progression, and anti-cancer drug screening with unprecedented control and reproducibility. In this review, we discuss the role of 3D bioprinting in reconstituting oral cancer, the prospects of application to fill the literature gap, and the challenges that need to be addressed in order to exploit this emerging technology for future work in oral cancer research.
Collapse
Affiliation(s)
- Thafar Almela
- College of Dentistry, University of Mosul, Mosul 41002, Iraq
| | - Lobat Tayebi
- School of Dentistry, Marquette University, Milwaukee, WI 53233, USA
| | - Keyvan Moharamzadeh
- Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
37
|
Metzger W, Rösch B, Sossong D, Bubel M, Pohlemann T. Flow cytometric quantification of apoptotic and proliferating cells applying an improved method for dissociation of spheroids. Cell Biol Int 2021; 45:1633-1643. [PMID: 33913594 DOI: 10.1002/cbin.11618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/30/2021] [Accepted: 04/18/2021] [Indexed: 01/31/2023]
Abstract
Spheroids are a promising tool for many cell culture applications, but their microscopic analysis is limited. Flow cytometry on a single cell basis, which requires a gentle but also efficient dissociation of spheroids, could be an alternative analysis. Mono-culture and coculture spheroids consisting of human fibroblasts and human endothelial cells were generated by the liquid overlay technique and were dissociated using AccuMax as a dissociation agent combined with gentle mechanical forces. This study aimed to quantify the number of apoptotic and proliferative cells. We were able to dissociate spheroids of differing size, age, and cellular composition in a single-step dissociation protocol within 10 min. The number of single cells was higher than 95% and in most cases, the viability of the cells after dissociation was higher than 85%. Coculture spheroids exhibited a higher sensitivity as shown by lower viability, higher amount of cellular debris, and a higher amount of apoptotic cells. Considerable expression of the proliferation marker Ki67 could only be seen in 1-day-old spheroids but was already downregulated on Day 3. In summary, our dissociation protocol enabled a fast and gentle dissociation of spheroids for the subsequent flow cytometric analysis. The chosen cell type had a strong influence on cell viability and apoptosis. Initially high rates of proliferative cells decreased rapidly and reached values of healthy tissue 3 days after generation of the spheroids. In conclusion, the flow cytometry of dissociated spheroids could be a promising analytical tool, which could be ideally combined with microscopic techniques.
Collapse
Affiliation(s)
- Wolfgang Metzger
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Barbara Rösch
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Daniela Sossong
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Monika Bubel
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Tim Pohlemann
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
38
|
Augustine R, Kalva SN, Ahmad R, Zahid AA, Hasan S, Nayeem A, McClements L, Hasan A. 3D Bioprinted cancer models: Revolutionizing personalized cancer therapy. Transl Oncol 2021; 14:101015. [PMID: 33493799 PMCID: PMC7823217 DOI: 10.1016/j.tranon.2021.101015] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
After cardiovascular disease, cancer is the leading cause of death worldwide with devastating health and economic consequences, particularly in developing countries. Inter-patient variations in anti-cancer drug responses further limit the success of therapeutic interventions. Therefore, personalized medicines approach is key for this patient group involving molecular and genetic screening and appropriate stratification of patients to treatment regimen that they will respond to. However, the knowledge related to adequate risk stratification methods identifying patients who will respond to specific anti-cancer agents is still lacking in many cancer types. Recent advancements in three-dimensional (3D) bioprinting technology, have been extensively used to generate representative bioengineered tumor in vitro models, which recapitulate the human tumor tissues and microenvironment for high-throughput drug screening. Bioprinting process involves the precise deposition of multiple layers of different cell types in combination with biomaterials capable of generating 3D bioengineered tissues based on a computer-aided design. Bioprinted cancer models containing patient-derived cancer and stromal cells together with genetic material, extracellular matrix proteins and growth factors, represent a promising approach for personalized cancer therapy screening. Both natural and synthetic biopolymers have been utilized to support the proliferation of cells and biological material within the personalized tumor models/implants. These models can provide a physiologically pertinent cell-cell and cell-matrix interactions by mimicking the 3D heterogeneity of real tumors. Here, we reviewed the potential applications of 3D bioprinted tumor constructs as personalized in vitro models in anticancer drug screening and in the establishment of precision treatment regimens.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar.
| | - Sumama Nuthana Kalva
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Rashid Ahmad
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Alap Ali Zahid
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Shajia Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Ajisha Nayeem
- Department of Biotechnology, St. Mary's College, Thrissur, 680020, Kerala, India
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, 2007, NSW, Australia
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar.
| |
Collapse
|
39
|
Katti KS, Jasuja H, Kar S, Katti DR. Nanostructured Biomaterials for In Vitro Models of Bone Metastasis Cancer. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 17:100254. [PMID: 33718691 PMCID: PMC7948119 DOI: 10.1016/j.cobme.2020.100254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In recent years, tissue engineering approaches have attracted substantial attention owing to their ability to create physiologically relevant in vitro disease models that closely mimic in vivo conditions. Here, we review nanocomposite materials and scaffolds used for the design of in vitro models of cancer, including metastatic sites. We discuss the role of material properties in modulating cellular phenotype in 3D disease models. Also, we highlight the application of tissue-engineered bone as a tool for faithful recapitulation of the microenvironment of metastatic prostate and breast cancer, since these two types of cancer have the propensity to metastasize to bone. Overall, we summarize recent efforts on developing 3D in vitro models of bone metastatic cancers that provide a platform to study tumor progression and facilitate high-throughput drug screening.
Collapse
Affiliation(s)
- Kalpana S. Katti
- Center for Engineered Cancer Test Beds, Department of Civil and Environmental Engineering North Dakota State University, Fargo ND 58108, USA
| | - Haneesh Jasuja
- Center for Engineered Cancer Test Beds, Department of Civil and Environmental Engineering North Dakota State University, Fargo ND 58108, USA
| | - Sumanta Kar
- Center for Engineered Cancer Test Beds, Department of Civil and Environmental Engineering North Dakota State University, Fargo ND 58108, USA
| | - Dinesh R. Katti
- Center for Engineered Cancer Test Beds, Department of Civil and Environmental Engineering North Dakota State University, Fargo ND 58108, USA
| |
Collapse
|
40
|
Doppler imaging detects bacterial infection of living tissue. Commun Biol 2021; 4:178. [PMID: 33568744 PMCID: PMC7876006 DOI: 10.1038/s42003-020-01550-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 11/25/2020] [Indexed: 01/17/2023] Open
Abstract
Living 3D in vitro tissue cultures, grown from immortalized cell lines, act as living sentinels as pathogenic bacteria invade the tissue. The infection is reported through changes in the intracellular dynamics of the sentinel cells caused by the disruption of normal cellular function by the infecting bacteria. Here, the Doppler imaging of infected sentinels shows the dynamic characteristics of infections. Invasive Salmonella enterica serovar Enteritidis and Listeria monocytogenes penetrate through multicellular tumor spheroids, while non-invasive strains of Escherichia coli and Listeria innocua remain isolated outside the cells, generating different Doppler signatures. Phase distributions caused by intracellular transport display Lévy statistics, introducing a Lévy-alpha spectroscopy of bacterial invasion. Antibiotic treatment of infected spheroids, monitored through time-dependent Doppler shifts, can distinguish drug-resistant relative to non-resistant strains. This use of intracellular Doppler spectroscopy of living tissue sentinels opens a new class of microbial assay with potential importance for studying the emergence of antibiotic resistance. Honggu Choi et al. use biodynamic Doppler imaging to monitor bacterial infection of 3D living tissue and describe changes in the intracellular motions of living host tissue induced by early-stage infection. This work demonstrates the potential for the clinical use of this method to test for antibiotic-resistant infections.
Collapse
|
41
|
Kulwatno J, Gearhart J, Gong X, Herzog N, Getzin M, Skobe M, Mills KL. Growth of tumor emboli within a vessel model reveals dependence on the magnitude of mechanical constraint. Integr Biol (Camb) 2021; 13:1-16. [PMID: 33443535 DOI: 10.1093/intbio/zyaa024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/02/2020] [Accepted: 12/03/2020] [Indexed: 01/18/2023]
Abstract
Tumor emboli-aggregates of tumor cells within vessels-pose a clinical challenge as they are associated with increased metastasis and tumor recurrence. When growing within a vessel, tumor emboli are subject to a unique mechanical constraint provided by the tubular geometry of the vessel. Current models of tumor emboli use unconstrained multicellular tumor spheroids, which neglect this mechanical interplay. Here, we modeled a lymphatic vessel as a 200 μm-diameter channel in either a stiff or soft, bioinert agarose matrix to create a vessel-like constraint model (VLCM), and we modeled colon or breast cancer tumor emboli with aggregates of HCT116 or SUM149PT cells, respectively. The stiff matrix VLCM constrained the tumor emboli to the cylindrical channel, which led to continuous growth of the emboli, in contrast to the growth rate reduction that unconstrained spheroids exhibit. Emboli morphology in the soft matrix VLCM, however, was dependent on the magnitude of mechanical mismatch between the matrix and the cell aggregates. In general, when the elastic modulus of the matrix of the VLCM was greater than the emboli (EVLCM/Eemb > 1), the emboli were constrained to grow within the channel, and when the elastic modulus of the matrix was less than the emboli (0 < EVLCM/Eemb < 1), the emboli bulged into the matrix. Due to a large difference in myosin II expression between the cell lines, we hypothesized that tumor cell aggregate stiffness is an indicator of cellular force-generating capability. Inhibitors of myosin-related force generation decreased the elastic modulus and/or increased the stress relaxation of the tumor cell aggregates, effectively increasing the mechanical mismatch. The increased mechanical mismatch after drug treatment was correlated with increased confinement of tumor emboli growth along the channel, which may translate to increased tumor burden due to the increased tumor volume within the diffusion distance of nutrients and oxygen.
Collapse
Affiliation(s)
- Jonathan Kulwatno
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Jamie Gearhart
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Xiangyu Gong
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Nora Herzog
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Matthew Getzin
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Mihaela Skobe
- Department of Oncological Sciences & Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristen L Mills
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| |
Collapse
|
42
|
3D bioprinting of high cell-density heterogeneous tissue models through spheroid fusion within self-healing hydrogels. Nat Commun 2021; 12:753. [PMID: 33531489 PMCID: PMC7854667 DOI: 10.1038/s41467-021-21029-2] [Citation(s) in RCA: 265] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 01/07/2021] [Indexed: 02/06/2023] Open
Abstract
Cellular models are needed to study human development and disease in vitro, and to screen drugs for toxicity and efficacy. Current approaches are limited in the engineering of functional tissue models with requisite cell densities and heterogeneity to appropriately model cell and tissue behaviors. Here, we develop a bioprinting approach to transfer spheroids into self-healing support hydrogels at high resolution, which enables their patterning and fusion into high-cell density microtissues of prescribed spatial organization. As an example application, we bioprint induced pluripotent stem cell-derived cardiac microtissue models with spatially controlled cardiomyocyte and fibroblast cell ratios to replicate the structural and functional features of scarred cardiac tissue that arise following myocardial infarction, including reduced contractility and irregular electrical activity. The bioprinted in vitro model is combined with functional readouts to probe how various pro-regenerative microRNA treatment regimes influence tissue regeneration and recovery of function as a result of cardiomyocyte proliferation. This method is useful for a range of biomedical applications, including the development of precision models to mimic diseases and the screening of drugs, particularly where high cell densities and heterogeneity are important.
Collapse
|
43
|
Advanced Multi-Dimensional Cellular Models as Emerging Reality to Reproduce In Vitro the Human Body Complexity. Int J Mol Sci 2021; 22:ijms22031195. [PMID: 33530487 PMCID: PMC7865724 DOI: 10.3390/ijms22031195] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
A hot topic in biomedical science is the implementation of more predictive in vitro models of human tissues to significantly improve the knowledge of physiological or pathological process, drugs discovery and screening. Bidimensional (2D) culture systems still represent good high-throughput options for basic research. Unfortunately, these systems are not able to recapitulate the in vivo three-dimensional (3D) environment of native tissues, resulting in a poor in vitro–in vivo translation. In addition, intra-species differences limited the use of animal data for predicting human responses, increasing in vivo preclinical failures and ethical concerns. Dealing with these challenges, in vitro 3D technological approaches were recently bioengineered as promising platforms able to closely capture the complexity of in vivo normal/pathological tissues. Potentially, such systems could resemble tissue-specific extracellular matrix (ECM), cell–cell and cell–ECM interactions and specific cell biological responses to mechanical and physical/chemical properties of the matrix. In this context, this review presents the state of the art of the most advanced progresses of the last years. A special attention to the emerging technologies for the development of human 3D disease-relevant and physiological models, varying from cell self-assembly (i.e., multicellular spheroids and organoids) to the use of biomaterials and microfluidic devices has been given.
Collapse
|
44
|
Human Embryo Models and Drug Discovery. Int J Mol Sci 2021; 22:ijms22020637. [PMID: 33440617 PMCID: PMC7828037 DOI: 10.3390/ijms22020637] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
For obvious reasons, such as, e.g., ethical concerns or sample accessibility, model systems are of highest importance to study the underlying molecular mechanisms of human maladies with the aim to develop innovative and effective therapeutic strategies. Since many years, animal models and highly proliferative transformed cell lines are successfully used for disease modelling, drug discovery, target validation, and preclinical testing. Still, species-specific differences regarding genetics and physiology and the limited suitability of immortalized cell lines to draw conclusions on normal human cells or specific cell types, are undeniable shortcomings. The progress in human pluripotent stem cell research now allows the growth of a virtually limitless supply of normal and DNA-edited human cells, which can be differentiated into various specific cell types. However, cells in the human body never fulfill their functions in mono-lineage isolation and diseases always develop in complex multicellular ecosystems. The recent advances in stem cell-based 3D organoid technologies allow a more accurate in vitro recapitulation of human pathologies. Embryoids are a specific type of such multicellular structures that do not only mimic a single organ or tissue, but the entire human conceptus or at least relevant components of it. Here we briefly describe the currently existing in vitro human embryo models and discuss their putative future relevance for disease modelling and drug discovery.
Collapse
|
45
|
Brown MJ, Morris MA, Akam EC. An exploration of the role of exercise in modulating breast cancer progression in vitro: a systematic review and meta-analysis. Am J Physiol Cell Physiol 2020; 320:C253-C263. [PMID: 33356943 DOI: 10.1152/ajpcell.00461.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Breast cancer is the most prevalent cancer in women worldwide. In the United Kingdom, approximately 5% of all breast cancers are already metastatic at the time of diagnosis. An abundance of literature shows that exercise can have beneficial effects on the outcome and prognosis of breast cancer patients, yet the molecular mechanisms remain poorly understood. There are several in vitro models that aim to recapitulate the response of breast cancer to exercise in vivo; this systematic review and meta-analysis summarizes the existing literature. The following search terms were used to conduct a systematic literature search using a collection of databases (last search performed May 2020): "in vitro," "exercise," and "breast cancer." Only studies that investigated the effects of exercise on breast cancer in vitro were included. Standardized mean differences (SMD) were calculated to determine pooled effect sizes. This meta-analysis has successfully demonstrated that various identified exercise interventions on breast cancer cells in vitro significantly reduced breast cancer cell viability, proliferation, and tumorigenic potential (SMD = -1.76, P = 0.004, SMD = -2.85, P = 0.003, and SMD = -3.15, P = 0.0008, respectively). A clear direction of effect was found with exercise on breast cancer cell migration in vitro, however this effect was not significant (SMD = -0.62, P = 0.317). To our knowledge, this is the first meta-analysis and systematic review investigating and summarizing literature on exercise and breast cancer in vitro, highlighting models used and priority areas for future research focus.
Collapse
Affiliation(s)
- Marie-Juliet Brown
- School of Sports, Exercise and Health Sciences, Loughborough University, Leicestershire, United Kingdom
| | - Mhairi A Morris
- School of Sports, Exercise and Health Sciences, Loughborough University, Leicestershire, United Kingdom
| | - Elizabeth C Akam
- School of Sports, Exercise and Health Sciences, Loughborough University, Leicestershire, United Kingdom
| |
Collapse
|
46
|
Functional Screening Techniques to Identify Long Non-Coding RNAs as Therapeutic Targets in Cancer. Cancers (Basel) 2020; 12:cancers12123695. [PMID: 33317042 PMCID: PMC7763270 DOI: 10.3390/cancers12123695] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Long non-coding RNAs (lncRNAs) are a recently discovered class of molecules in the cell, with potential to be utilized as therapeutic targets in cancer. A number of lncRNAs have been described to play important roles in tumor progression and drive molecular processes involved in cell proliferation, apoptosis or invasion. However, the vast majority of lncRNAs have not been studied in the context of cancer thus far. With the advent of CRISPR/Cas genome editing, high-throughput functional screening approaches to identify lncRNAs that impact cancer growth are becoming more accessible. Here, we review currently available methods to study hundreds to thousands of lncRNAs in parallel to elucidate their role in tumorigenesis and cancer progression. Abstract Recent technological advancements such as CRISPR/Cas-based systems enable multiplexed, high-throughput screening for new therapeutic targets in cancer. While numerous functional screens have been performed on protein-coding genes to date, long non-coding RNAs (lncRNAs) represent an emerging class of potential oncogenes and tumor suppressors, with only a handful of large-scale screens performed thus far. Here, we review in detail currently available screening approaches to identify new lncRNA drivers of tumorigenesis and tumor progression. We discuss the various approaches of genomic and transcriptional targeting using CRISPR/Cas9, as well as methods to post-transcriptionally target lncRNAs via RNA interference (RNAi), antisense oligonucleotides (ASOs) and CRISPR/Cas13. We discuss potential advantages, caveats and future applications of each method to provide an overview and guide on investigating lncRNAs as new therapeutic targets in cancer.
Collapse
|
47
|
Addressing the tumour microenvironment in early drug discovery: a strategy to overcome drug resistance and identify novel targets for cancer therapy. Drug Discov Today 2020; 26:663-676. [PMID: 33278601 DOI: 10.1016/j.drudis.2020.11.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/04/2020] [Accepted: 11/26/2020] [Indexed: 12/20/2022]
Abstract
The tumour microenvironment (TME) comprises not only malignant and non-malignant cells, but also the extracellular matrix (ECM), secreted factors, and regulators of cellular functions. In addition to genetic alterations, changes of the biochemical/biophysical properties or cellular composition of the TME have been implicated in drug resistance. Here, we review the composition of the ECM and different elements of the TME contributing to drug resistance, including soluble factors, hypoxia, extracellular acidity, and cell adhesion properties. We discuss selected approaches for modelling the TME, current progress, and their use in low-and high-throughput assays for preclinical studies. Lastly, we summarise the status quo of advanced 3D cancer models compatible with high-throughput screening (HTS), the technical practicalities and challenges.
Collapse
|
48
|
Craig M, Jenner AL, Namgung B, Lee LP, Goldman A. Engineering in Medicine To Address the Challenge of Cancer Drug Resistance: From Micro- and Nanotechnologies to Computational and Mathematical Modeling. Chem Rev 2020; 121:3352-3389. [PMID: 33152247 DOI: 10.1021/acs.chemrev.0c00356] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drug resistance has profoundly limited the success of cancer treatment, driving relapse, metastasis, and mortality. Nearly all anticancer drugs and even novel immunotherapies, which recalibrate the immune system for tumor recognition and destruction, have succumbed to resistance development. Engineers have emerged across mechanical, physical, chemical, mathematical, and biological disciplines to address the challenge of drug resistance using a combination of interdisciplinary tools and skill sets. This review explores the developing, complex, and under-recognized role of engineering in medicine to address the multitude of challenges in cancer drug resistance. Looking through the "lens" of intrinsic, extrinsic, and drug-induced resistance (also referred to as "tolerance"), we will discuss three specific areas where active innovation is driving novel treatment paradigms: (1) nanotechnology, which has revolutionized drug delivery in desmoplastic tissues, harnessing physiochemical characteristics to destroy tumors through photothermal therapy and rationally designed nanostructures to circumvent cancer immunotherapy failures, (2) bioengineered tumor models, which have benefitted from microfluidics and mechanical engineering, creating a paradigm shift in physiologically relevant environments to predict clinical refractoriness and enabling platforms for screening drug combinations to thwart resistance at the individual patient level, and (3) computational and mathematical modeling, which blends in silico simulations with molecular and evolutionary principles to map mutational patterns and model interactions between cells that promote resistance. On the basis that engineering in medicine has resulted in discoveries in resistance biology and successfully translated to clinical strategies that improve outcomes, we suggest the proliferation of multidisciplinary science that embraces engineering.
Collapse
Affiliation(s)
- Morgan Craig
- Department of Mathematics and Statistics, University of Montreal, Montreal, Quebec H3C 3J7, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, Quebec H3S 2G4, Canada
| | - Adrianne L Jenner
- Department of Mathematics and Statistics, University of Montreal, Montreal, Quebec H3C 3J7, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, Quebec H3S 2G4, Canada
| | - Bumseok Namgung
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02139, United States
| | - Luke P Lee
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02139, United States
| | - Aaron Goldman
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02139, United States
| |
Collapse
|
49
|
Frieboes HB, Raghavan S, Godin B. Modeling of Nanotherapy Response as a Function of the Tumor Microenvironment: Focus on Liver Metastasis. Front Bioeng Biotechnol 2020; 8:1011. [PMID: 32974325 PMCID: PMC7466654 DOI: 10.3389/fbioe.2020.01011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment (TME) presents a challenging barrier for effective nanotherapy-mediated drug delivery to solid tumors. In particular for tumors less vascularized than the surrounding normal tissue, as in liver metastases, the structure of the organ itself conjures with cancer-specific behavior to impair drug transport and uptake by cancer cells. Cells and elements in the TME of hypovascularized tumors play a key role in the process of delivery and retention of anti-cancer therapeutics by nanocarriers. This brief review describes the drug transport challenges and how they are being addressed with advanced in vitro 3D tissue models as well as with in silico mathematical modeling. This modeling complements network-oriented techniques, which seek to interpret intra-cellular relevant pathways and signal transduction within cells and with their surrounding microenvironment. With a concerted effort integrating experimental observations with computational analyses spanning from the molecular- to the tissue-scale, the goal of effective nanotherapy customized to patient tumor-specific conditions may be finally realized.
Collapse
Affiliation(s)
- Hermann B. Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, United States
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
- Center for Predictive Medicine, University of Louisville, Louisville, KY, United States
| | - Shreya Raghavan
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, United States
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
- Department of Obstetrics and Gynecology, Houston Methodist Hospital, Houston, TX, United States
- Developmental Therapeutics Program, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
50
|
Kock A, Bergqvist F, Steinmetz J, Elfman LHM, Korotkova M, Johnsen JI, Jakobsson PJ, Kogner P, Larsson K. Establishment of an in vitro 3D model for neuroblastoma enables preclinical investigation of combined tumor-stroma drug targeting. FASEB J 2020; 34:11101-11114. [PMID: 32623799 DOI: 10.1096/fj.202000684r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/15/2022]
Abstract
The majority of anti-cancer therapies target the proliferating tumor cells, while the tumor stroma, principally unaffected, survives, and provide a niche for surviving tumor cells. Combining tumor cell and stroma-targeting therapies thus have a potential to improve patient outcome. The neuroblastoma stroma contains cancer-associated fibroblasts expressing microsomal prostaglandin E synthase-1 (mPGES-1). mPGES-1-derived prostaglandin E2 (PGE2 ) is known to promote tumor growth through increased proliferation and survival of tumor cells, immune suppression, angiogenesis, and therapy resistance, and we, therefore, hypothesize that mPGES-1 constitutes an interesting stromal target. Here, we aimed to develop a relevant in vitro model to study combination therapies. Co-culturing of neuroblastoma and fibroblast cells in 3D tumor spheroids mimic neuroblastoma tumors with regard to the cyclooxygenase/mPGES-1/PGE2 pathway. Using the spheroid model, we show that the inhibition of fibroblast-derived mPGES-1 enhanced the cytotoxic effect of doxorubicin and vincristine and significantly reduced tumor cell viability and spheroid growth. Cyclic treatment with vincristine in combination with an mPGES-1 inhibitor abrogated cell repopulation. Moreover, inhibition of mPGES-1 potentiated the cytotoxic effect of vincristine on established neuroblastoma allografts in mice. In conclusion, we established a 3D neuroblastoma model, highlighting the potential of combining stromal targeting of mPGES-1 with tumor cell targeting drugs like vincristine.
Collapse
Affiliation(s)
- Anna Kock
- Childhood Cancer Research Unit, Department of Children's and Women's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Filip Bergqvist
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Julia Steinmetz
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lotta H M Elfman
- Childhood Cancer Research Unit, Department of Children's and Women's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Marina Korotkova
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Children's and Women's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Children's and Women's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Karin Larsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|