1
|
Vasudevan J, Vijayakumar R, Reales-Calderon JA, Lam MSY, Ow JR, Aw J, Tan D, Tan AT, Bertoletti A, Adriani G, Pavesi A. In vitro integration of a functional vasculature to model endothelial regulation of chemotherapy and T-cell immunotherapy in liver cancer. Biomaterials 2025; 320:123175. [PMID: 40043483 DOI: 10.1016/j.biomaterials.2025.123175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 04/06/2025]
Abstract
The complex tumor microenvironment (TME) presents significant challenges to the development of effective therapies against solid tumors, highlighting the need for advanced in vitro models that better recapitulate TME biology. To address this, we developed a vascularized human liver tumor model using a microfluidic platform, designed to test both drug and cell-based therapies. This model mimics critical tumorigenic features such as hypoxia, extracellular matrix (ECM), and perfusable vascular networks. Intravascular administration of Sorafenib demonstrated its ability to disrupt vascular structures significantly, while eliciting heterogeneous responses in two distinct liver tumor cell lines, HepG2 and Hep3b. Furthermore, treatment with engineered T-cells revealed that the tumor vasculature impeded T-cell infiltration into the tumor core but preserved their cytotoxic capacity, albeit with reduced exhaustion levels. Cytokine analysis and spatial profiling of vascularized tumor samples identified proinflammatory factors that may enhance T-cell-mediated antitumor responses. By capturing key TME characteristics, this microfluidic platform provides a powerful tool enabling detailed investigation of tumor-immune and tumor-vascular interactions. Its versatility could serve as a promising bridge between preclinical studies and clinical testing, offering opportunities for developing and optimizing personalized therapeutic strategies for solid tumors.
Collapse
Affiliation(s)
- Jyothsna Vasudevan
- Mechanobiology Institute, National University of Singapore (NUS), 5A Engineering Drive 1, Singapore, 117411, Republic of Singapore
| | - Ragavi Vijayakumar
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Jose Antonio Reales-Calderon
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Maxine S Y Lam
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Jin Rong Ow
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Joey Aw
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Damien Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Anthony Tanoto Tan
- Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore
| | - Antonio Bertoletti
- Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore
| | - Giulia Adriani
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A∗STAR), 8A Biomedical Grove, Immunos, Singapore, 138648, Republic of Singapore; Department of Biomedical Engineering, National University of Singapore (NUS), 4 Engineering Drive 3, Singapore, 117583, Republic of Singapore
| | - Andrea Pavesi
- Mechanobiology Institute, National University of Singapore (NUS), 5A Engineering Drive 1, Singapore, 117411, Republic of Singapore; Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore; Lee Kong Chian School of Medicine (LKCMedicine), Cancer Discovery and Regenerative Medicine Program, Nanyang Technological University, 308232, Republic of Singapore.
| |
Collapse
|
2
|
Shin S, Choi Y, Jang W, Ulziituya B, Ha G, Kang R, Park S, Kim M, Zhang YS, Kim HJ, Lee J. A vascularized tumors-on-a-chip model for studying tumor-angiogenesis interplay, heterogeneity and drug responses. Mater Today Bio 2025; 32:101741. [PMID: 40275956 PMCID: PMC12020855 DOI: 10.1016/j.mtbio.2025.101741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/26/2025] Open
Abstract
Current tumor models struggle to replicate the complexity of the tumor microenvironment, particularly endothelial sprouting and vascular-tumor interactions. To address these limitations, we developed a vascularized tumors-on-a-chip model by fusing tumor spheroids with HUVEC spheroids to simulate angiogenesis. The model incorporates hypoxia-driven cytokine secretion and dynamic endothelial penetration, enabling accurate recapitulation of angiogenic processes. Spheroids were optimized for size and viability, and four cancer types were studied, with GBM and A549 exhibiting the highest angiogenic potential, as confirmed by Z-stack imaging and qRT-PCR. Encapsulation in GelMA and integration into PDMS-based microfluidic chips provided a dynamic flow environment, mimicking in vivo drug delivery while enabling high-throughput drug screening. This chip-based system allows simultaneous testing of multiple drugs or tumors under physiologically relevant conditions, enhancing its translational potential. The platform was validated using doxorubicin and bevacizumab, revealing reduced VEGF secretion and dynamic cytokine responses, replicating vascular barriers. Further validation in murine models demonstrated its capacity to promote angiogenesis and mimic tumor-vessel interactions. This advanced tumors-on-a-chip model addresses critical shortcomings of conventional 2D and 3D systems and offers a transformative tool for preclinical drug evaluation and the development of precision oncology strategies, bridging the gap between in vitro testing and in vivo relevance.
Collapse
Affiliation(s)
- Suyeon Shin
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
| | - Yurim Choi
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
| | - WonJun Jang
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | | | - Giheon Ha
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
| | - Raehui Kang
- Division of Interdisciplinary Bioscience & Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
| | - Soojin Park
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Minseok Kim
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, MA, USA
| | - Han-Jun Kim
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Junmin Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 790-784, Republic of Korea
| |
Collapse
|
3
|
Maity S, Jewell C, Yilgor C, Kawakita S, Sharma S, Gomez A, Mecwan M, Falcone N, Ermis M, Monirizad M, Kouchehbaghi NH, Zehtabi F, Khorsandi D, Dokmeci MR, Moniz-Garcia D, Quiñones-Hinojosa A, Khademhosseini A, Jucaud V. Deciphering pericyte-induced temozolomide resistance in glioblastoma with a 3D microphysiological system mimicking the biomechanical properties of brain tissue. Acta Biomater 2025:S1742-7061(25)00363-0. [PMID: 40383349 DOI: 10.1016/j.actbio.2025.05.038] [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: 11/27/2024] [Revised: 04/29/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Glioblastoma (GBM) is a highly aggressive malignancy with a poor prognosis and frequent resistance to temozolomide (TMZ), the standard-of-care chemotherapy. The complex mechanisms underlying GBM chemoresistance, particularly the role of pericytes, remain poorly understood due to the lack of physiologically relevant in vitro models replicating the complex tumor microenvironment (TME). Here, we present a biomimetic 3D GBM microphysiological system that replicates the biomechanical properties of brain tissue (G'∼800Pa, G"∼100Pa) and enables the study of pericyte-mediated TMZ resistance. GBM spheroids (U87, LN229, PDM140) were cultured alone or co-cultured with pericytes in a composite hydrogel for 14 days and remained viable and proliferative. In response to TMZ, PDM140 was the most sensitive (IC50=73μM), followed by LN229 (IC50=278μM) and U87 (IC50=446μM). Co-culture with pericytes significantly increased GBM spheroid viability by 22.7% (PDM140), 32.5% (LN229), and 22.1% (U87), confirming pericyte-induced TMZ resistance. Notably, pericytes exhibited a 160-fold upregulation of C-C motif chemokine ligand 5 (CCL5) upon TMZ treatment, implicating the CCL5-mediated pathway in chemoresistance. This innovative brain-mimicking 3D GBM model provides a physiologically relevant platform for studying tumor-pericyte interactions and testing therapeutic strategies targeting CCL5-mediated resistance mechanisms in GBM. STATEMENT OF SIGNIFICANCE: We developed a multicellular 3D glioblastoma microphysiological system mimicking the physicochemical properties of brain tissues and pericyte-mediated TMZ resistance that can be used to screen for standard-of-care chemotherapy. This advanced hydrogel-based platform demonstrated the critical role of the glioblastoma tumor microenvironment in modulating chemotherapy sensitivity, particularly the pericyte-induced CCL5-CCR5 paracrine axis that can lead to the identification of therapeutic targets within the CCL5-CCR5 pathway toward more effective treatments disrupting these resistance mechanisms. Overall, the proposed 3D glioblastoma microphysiological system can transform drug screening and personalized treatment for GBM by offering ethical and cost-effective alternatives to animal testing and more effective drug screening and discovery efforts, ultimately improving GBM patient outcomes.
Collapse
Affiliation(s)
- Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA.; Department of Orthopedic Surgery, Duke University School of Medicine, Duke University, Durham, NC 27705, USA
| | - Christopher Jewell
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Can Yilgor
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Saurabh Sharma
- Department of Surgery, Stanford School of Medicine, Stanford University Medical Center, CA 94305, USA
| | - Alejandro Gomez
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Mahsa Monirizad
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | | | - Fatemeh Zehtabi
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | | | - Diogo Moniz-Garcia
- Mayo Clinic Florida, Department of Neurosurgery, Jacksonville, FL, 32224, USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA..
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA..
| |
Collapse
|
4
|
Jonckheere S, Taminau J, Adams J, Haerinck J, De Coninck J, Verstappe J, De Clercq K, Peeters E, Gheldof A, De Smedt E, Goossens V, Audenaert D, Candi A, Versele M, De Groote D, Verschuere H, Stemmler M, Brabletz T, Vandenabeele P, Casali A, Campbell K, Goossens S, Berx G. Development and validation of a high-throughput screening pipeline of compound libraries to target EMT. Cell Death Differ 2025:10.1038/s41418-025-01515-6. [PMID: 40341726 DOI: 10.1038/s41418-025-01515-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 03/18/2025] [Accepted: 04/08/2025] [Indexed: 05/11/2025] Open
Abstract
Epithelial to Mesenchymal transitions (EMT) drive cell plasticity and are associated with cell features such as invasiveness, migration and stemness. They are orchestrated by select families of EMT-associated transcription factors, which exhibit pleiotropic roles in the malignant progression of various cancer types, such as breast and colorectal cancer (CRC). This has spurred interest in EMT as a promising target for the development of novel therapeutic strategies. In this study, we developed a phenotypic dual EMT Sensor screening assay, amendable to efficient high-throughput identification of small molecules interfering with EMT. In a proof-of-concept screening we identified anti-EMT repurposing drugs. From these, we validated RepSox, a selective inhibitor of the TGF-β type I receptor ALK5, and demonstrated that it is potently blocking EMT in both breast and colorectal cancer cell lines in vitro. In addition, utilizing a Drosophila melanogaster metastatic CRC model we confirmed the ability of the identified anti-EMT hits to suppress metastatic behavior in vivo.
Collapse
Affiliation(s)
- Sven Jonckheere
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Molecular and Cellular Oncology Laboratory, Ghent University, Ghent, Belgium
| | - Joachim Taminau
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Molecular and Cellular Oncology Laboratory, Ghent University, Ghent, Belgium
| | - Jamie Adams
- Department of Biomedical Science, The University of Sheffield, Sheffield, UK
| | - Jef Haerinck
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Molecular and Cellular Oncology Laboratory, Ghent University, Ghent, Belgium
| | - Jordy De Coninck
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Molecular and Cellular Oncology Laboratory, Ghent University, Ghent, Belgium
| | - Jeroen Verstappe
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Molecular and Cellular Oncology Laboratory, Ghent University, Ghent, Belgium
| | - Kato De Clercq
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Molecular and Cellular Oncology Laboratory, Ghent University, Ghent, Belgium
| | - Evelien Peeters
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Molecular and Cellular Oncology Laboratory, Ghent University, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Alexander Gheldof
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Molecular and Cellular Oncology Laboratory, Ghent University, Ghent, Belgium
- Centre for Medical Genetics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Eva De Smedt
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Molecular and Cellular Oncology Laboratory, Ghent University, Ghent, Belgium
| | - Vera Goossens
- VIB Screening Core & UGent Expertise Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - Dominique Audenaert
- VIB Screening Core & UGent Expertise Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - Aurélie Candi
- Cistim Leuven vzw & Centre for Drug Design and Discovery (CD3), KU Leuven, Leuven, Belgium
| | - Matthias Versele
- Cistim Leuven vzw & Centre for Drug Design and Discovery (CD3), KU Leuven, Leuven, Belgium
| | - Dominic De Groote
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Hanne Verschuere
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Molecular and Cellular Oncology Laboratory, Ghent University, Ghent, Belgium
- Cell Death and Inflammation Unit, VIB Center for Inflammation Research, Ghent, Belgium
| | - Marc Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Peter Vandenabeele
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Cell Death and Inflammation Unit, VIB Center for Inflammation Research, Ghent, Belgium
| | - Andreu Casali
- Department de Ciènces Mèdiques Bàsiques, Universitat de Llieda and IRBLleida, Lleida, Spain
| | - Kyra Campbell
- Department of Biomedical Science, The University of Sheffield, Sheffield, UK
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Geert Berx
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
- Molecular and Cellular Oncology Laboratory, Ghent University, Ghent, Belgium.
| |
Collapse
|
5
|
Liu Z, Mao X, Xie Y, Yan Y, Wang X, Mi J, Yuan H, Zhang J, Huang C, Chen J, Jili M, Huang S, Zhang Q, Wang F, Mo Z, Yang R. Single-cell RNA sequencing reveals a fibroblast gene signature that promotes T-cell infiltration in muscle-invasive bladder cancer. Commun Biol 2025; 8:696. [PMID: 40319103 PMCID: PMC12049545 DOI: 10.1038/s42003-025-08094-9] [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: 06/14/2024] [Accepted: 04/16/2025] [Indexed: 05/07/2025] Open
Abstract
Muscle-invasive bladder cancer (MIBC) is characterized by a complex tumor microenvironment (TME) that drives aggressive progression and treatment resistance. Previous studies have highlighted the roles of cancer-associated fibroblasts (CAFs) and exhausted T (Tex) cells in MIBC, but their interactive mechanisms remain poorly understood. Here, single-cell RNA sequencing of 19 tissue samples from 12 patients-7 MIBC, 3 non-muscle-invasive bladder cancer (NMIBC), and 9 normal tissue samples-identified 13 transcriptionally distinct fibroblast clusters and 10 functionally heterogeneous T-cell subsets. Two interferon (IFN)-responsive fibroblast populations, F-ISG15 (inflammatory CAFs) and F-POSTN (myofibroblastic CAFs), were shown to predominate in the MIBC TME. In vivo experiments demonstrated that IFN-γ secreted by Tex cells polarizes CAFs to secrete CXCL12, which recruits CXCR4-expressing T cells via the CXCL12-CXCR4 chemotactic axis. Spatial analysis revealed a bidirectional loop: Tex-derived IFN-γ sustains CAF activation, whereas CAF-secreted CXCL12 amplifies Tex infiltration. Clinically, activated CAF signatures correlate with advanced disease stages and reduced patient survival in MIBC. These findings establish CXCL12 and IFN signaling as critical therapeutic targets, offering new strategies to disrupt immunosuppressive TME crosstalk and improve outcomes for MIBC patients.
Collapse
Affiliation(s)
- Zige Liu
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
| | - Xingning Mao
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Coconstructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuli Xie
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yunkun Yan
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiang Wang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
| | - Junhao Mi
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
| | - Hao Yuan
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiange Zhang
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Caisheng Huang
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The Nanning Second People's Hospital, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jianxin Chen
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Mujia Jili
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Shengzhu Huang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
| | - Qingyun Zhang
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Fubo Wang
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
| | - Zengnan Mo
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China.
| | - Rirong Yang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China.
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
6
|
Branco F, Cunha J, Mendes M, Sousa JJ, Vitorino C. 3D Bioprinting Models for Glioblastoma: From Scaffold Design to Therapeutic Application. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2501994. [PMID: 40116532 DOI: 10.1002/adma.202501994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Indexed: 03/23/2025]
Abstract
Conventional in vitro models fail to accurately mimic the tumor in vivo characteristics, being appointed as one of the causes of clinical attrition rate. Recent advances in 3D culture techniques, replicating essential physical and biochemical cues such as cell-cell and cell-extracellular matrix interactions, have led to the development of more realistic tumor models. Bioprinting has emerged to advance the creation of 3D in vitro models, providing enhanced flexibility, scalability, and reproducibility. This is crucial for the development of more effective drug treatments, and glioblastoma (GBM) is no exception. GBM, the most common and deadly brain cancer, remains a major challenge, with a median survival of only 15 months post-diagnosis. This review highlights the key components needed for 3D bioprinted GBM models. It encompasses an analysis of natural and synthetic biomaterials, along with crosslinking methods to improve structural integrity. Also, it critically evaluates current 3D bioprinted GBM models and their integration into GBM-on-a-chip platforms, which hold noteworthy potential for drug screening and personalized therapies. A versatile development framework grounded on Quality-by-Design principles is proposed to guide the design of bioprinting models. Future perspectives, including 4D bioprinting and machine learning approaches, are discussed, along with the current gaps to advance the field further.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| | - Joana Cunha
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| | - Maria Mendes
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| | - João J Sousa
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| |
Collapse
|
7
|
Ding R, Chen C, Wang L, Wang Y, Chai Z, He S, Zhang Q, Cheng S, Zou R. Matrix Stiffness Regulates the Osteogenic Differentiation of hPDLSCs via DNA Methylation. Int Dent J 2025; 75:100783. [PMID: 40315698 PMCID: PMC12090244 DOI: 10.1016/j.identj.2025.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 05/04/2025] Open
Abstract
OBJECTIVES This study aimed to examine the influence of matrix stiffness on osteogenic differentiation via epigenetic mechanisms in human periodontal ligament stem cells (hPDLSCs), with implications for understanding orthodontic tooth movement. MATERIALS AND METHODS hPDLSCs were cultured on substrates with varying stiffness (soft and stiff). Dot blot and immunofluorescence techniques were employed to measure global DNA methylation levels. RT-qPCR and alkaline phosphatase (ALP) activity assays were conducted to assess differences in DNA methylation and osteogenic potential. Additionally, ELISA was used to quantify DNA methyltransferase content and activity. RESULTS hPDLSCs on stiffer substrates exhibited increased 5-methylcytosine (5-mC) and higher global DNA methylation levels than those on soft substrates. With increased matrix stiffness, DNMT3A and DNMT3B mRNA expression levels rose. hPDLSCs on stiff matrices also showed elevated DNMT3B enzyme content and osteogenic activity. When global DNA methylation was reduced, mRNA levels of RUNX2, ALP, and Col-1 decreased, along with a notable reduction in ALP staining intensity in the inhibitor group. CONCLUSIONS Matrix stiffness is positively associated with global DNA methylation, with DNMT3B likely mediating this regulation in hPDLSCs. Furthermore, DNA methylation levels are positively linked to the osteogenic capability of hPDLSCs.
Collapse
Affiliation(s)
- Rong Ding
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Chen Chen
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Ling Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yijie Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Zhen Chai
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Siyu He
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Qianqian Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Shuli Cheng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| | - Rui Zou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| |
Collapse
|
8
|
Oh JM, Park Y, Lee J, Shen K. Microfabricated Organ-Specific Models of Tumor Microenvironments. Annu Rev Biomed Eng 2025; 27:307-333. [PMID: 40310890 DOI: 10.1146/annurev-bioeng-110222-103522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Despite the advances in detection, diagnosis, and treatments, cancer remains a lethal disease, claiming the lives of more than 600,000 people in the United States alone in 2024. To accelerate the development of new therapeutic strategies with improved responses, significant efforts have been made to develop microfabricated in vitro models of tumor microenvironments (TMEs) that address the limitations of animal-based cancer models. These models incorporate several advanced tissue engineering techniques to better reflect the organ- and patient-specific TMEs. Additionally, microfabricated models integrated with next-generation single-cell omics technologies provide unprecedented insights into patient's cellular and molecular heterogeneity and complexity. This review provides an overview of the recent understanding of cancer development and outlines the key TME elements that can be captured in microfabricated models to enhance their physiological relevance. We highlight the recent advances in microfabricated cancer models that reflect the unique characteristics of their organs of origin or sites of dissemination.
Collapse
Affiliation(s)
- Jeong Min Oh
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
| | - Yongkuk Park
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - Keyue Shen
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
9
|
Kah J, Staffeldt L, Volz T, Schulze K, Heumann A, Rövenstrunk G, Goebel M, Peine S, Dandri M, Lüth S. Classification of the LC4 Primarily-like Cell Line-Recapitulating a CDK4 Overexpressing Immune Evasive HIV-HCV-Induced HCC. Viruses 2025; 17:653. [PMID: 40431665 PMCID: PMC12115383 DOI: 10.3390/v17050653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality. HCC is characterized by high heterogeneity and, subsequently, adaptation by developing resistance to current treatments. Applying individualized models is crucial to understanding the potential of approved therapies. Therefore, we classify a primary-like cell line derived from the core region of an HCC with underlying HIV-HCV co-infection employing deep analysis on the pathway regulation level. METHODS We employed DEG analysis, followed by pathway analysis, to characterize the preservation level of the LC4 cells and the level of adoption. Next, we classify the new model for HCC research by employing healthy donor samples, commonly used HCC cell lines, and global RNAseq datasets. RESULTS LC4 cells reflect the characteristics of the parental cancer region, including immunosuppression and metabolic reprogramming, characterized by the downregulation of drug-metabolizing enzymes compared to healthy individuals, indicating a transition to alternate metabolic pathways. Moreover, we identified specific biomarkers equally regulated in the parental tissue, in global datasets of the same entities as well as in LC4 cells. CONCLUSIONS We classified LC4 cells as an individual immunosuppressive and highly progressive primary-like HCC cell line. LC4 cells are applicable as a model for preclinical drug testing, minimizing the lack of preclinical models in HCV-HIV-induced HCC research.
Collapse
Affiliation(s)
- Janine Kah
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany; (G.R.)
- Department of Gastroenterology, Center for Translational Medicine, University Hospital Brandenburg, 14770 Brandenburg, Germany
| | - Lisa Staffeldt
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany; (G.R.)
| | - Tassilo Volz
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
- German Center for Infection Research, Hamburg-Lübeck-Borstel Partner Site, 38124 Braunschweig, Germany
| | - Kornelius Schulze
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
| | - Asmus Heumann
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Götz Rövenstrunk
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany; (G.R.)
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Meike Goebel
- Institute for Transfusion Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sven Peine
- Institute for Transfusion Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maura Dandri
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
- German Center for Infection Research, Hamburg-Lübeck-Borstel Partner Site, 38124 Braunschweig, Germany
| | - Stefan Lüth
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany; (G.R.)
| |
Collapse
|
10
|
Zhang T, Yang L, Wang Z, Zhou N. SKOV-3 Cell Aggregates on a Microfluidic Chip with a Thermoresponsive Hydrogel as a Culture Scaffold for DOX Assessment. ACS OMEGA 2025; 10:14972-14979. [PMID: 40290958 PMCID: PMC12019744 DOI: 10.1021/acsomega.4c10301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/25/2025] [Accepted: 01/31/2025] [Indexed: 04/30/2025]
Abstract
Microfluidic chip technology is very popular in life sciences. Here, ovarian cancer SKOV-3 cell aggregates were formed using thermoresponsive poly(N-isopropylacrylamide-co-acrylic acid) (PNA) hydrogel as a culture scaffold on a microfluidic chip serving as an operating platform. A simple microfluidic chip was designed and fabricated as the three-dimensional (3D) cell culture microplatform. Different concentrations of doxorubicin (DOX) were fed to the obtained SKOV-3 cell aggregates on the chip via a pump. All characterization results indicated that this system could effectively perform 3D cell culture and drug evaluation to a certain extent. In addition, by grafting the RGD sequence, the biocompatibility of the PNA hydrogel was improved. On the one hand, the grafting of the RGD sequence into the hydrogel could significantly improve cell proliferation in this system; on the other hand, it led to an earlier appearance of DOX drug resistance. This versatile model in this study has the potential for further use in in vitro human ovarian cancer physiological models, drug discovery, and toxicology research.
Collapse
Affiliation(s)
- Tianzhu Zhang
- State Key Laboratory of Digital Medical
Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Liuxin Yang
- State Key Laboratory of Digital Medical
Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhengyang Wang
- State Key Laboratory of Digital Medical
Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Naizhen Zhou
- State Key Laboratory of Digital Medical
Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
11
|
Garre E, Rhost S, Gustafsson A, Szeponik L, Araujo TF, Quiding-Järbrink M, Helou K, Ståhlberg A, Landberg G. Breast cancer patient-derived scaffolds enhance the understanding of PD-L1 regulation and T cell cytotoxicity. Commun Biol 2025; 8:621. [PMID: 40240529 PMCID: PMC12003762 DOI: 10.1038/s42003-025-08054-3] [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: 01/08/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
Recent advances as well as obstacles for immune-based cancer treatment strategies, highlight the notable impact of patient cancer microenvironments on the immune cells and immune targets. Here, we use patient-derived scaffolds (PDS) generated from 110 primary breast cancers to monitor the impact of the cancer microenvironment on immune regulators. Pronounced variation in PD-L1 expression is observed in cancer cells adapted to different patient scaffolds. This variation is further linked to clinical observations and correlated with specific proteins detected in the cell-free PDSs using mass spectrometry. When adding T cells to the PDS-based cancer cultures, the killing efficiency of activated T cells vary between the cultures, whereas non-activated T cells modulate the cancer cell PD-L1 expression to treatment-predictive values, matching killing capacities of activated T cells. Surviving cancer cells show enrichment in cancer stem cell and epithelial-to-mesenchymal transition (EMT) features, suggesting that T cells may not efficiently target cells with metastatic potential. We conclude that clinically relevant insights in how to optimally target and guide immune-based cancer therapies can be obtained by including patient-derived scaffolds and cues from the cancer microenvironment in cancer patient handling and drug development.
Collapse
Affiliation(s)
- Elena Garre
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Sara Rhost
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
| | - Anna Gustafsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
| | - Louis Szeponik
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Thais Fenz Araujo
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
| | - Marianne Quiding-Järbrink
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Khalil Helou
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Ståhlberg
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Center for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Göran Landberg
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
12
|
Li C, Pan J, Shi Z, Zeng X, Xia X, He X, Wang W, Qiu B, Ding W, Huang D. Engineered Endometrial Clear Cell Cancer-on-a-Chip Reveals Early Invasion-Metastasis Cascade of Cancer Cells. Biomater Res 2025; 29:0177. [PMID: 40231208 PMCID: PMC11994883 DOI: 10.34133/bmr.0177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
Endometrial clear cell cancer (ECCC) is an extremely rare and highly malignant subtype of endometrial cancer. For most ECCC patients, cancer metastasis is the major cause of death. To date, due to the complexity of cancer evolution and the small number of cases, the metastasis of ECCC at the early stage remains largely unknown. Herein, we modeled the early invasion-metastasis cascade of ECCC by coculturing the ECCC patient-derived tumor cells (PDTCs) and primary human vascular endothelial cells on a microfluidic chip. With the chip, we for the first time replicated the dynamic migration of PDTCs into the surrounding stroma, including the intravasation and extravasation of PDTCs through the capillaries/microvessels, and presented the changes in the morphology and permeability of capillaries, with the decreased diameter and the increased permeability after cancer metastasis. We found that PDTCs were more invasive than the common endometrial adenocarcinoma cells. In addition, we preliminarily explored the inhibition of drugs on the early PDTC infiltration. This study provides new ideas for better understanding of ECCC evolution.
Collapse
Affiliation(s)
- Chengpan Li
- Department of Electronic Engineering and Information Science, School of Information Science and Technology,
University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Jing Pan
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Zhengdi Shi
- Department of Electronic Engineering and Information Science, School of Information Science and Technology,
University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xinyan Zeng
- Department of Integrated Traditional Chinese and Western Medicine,
Anhui Medical University, Hefei, Anhui 230032, China
| | - Xiaoping Xia
- Department of Obstetrics and Gynecology, Anhui Provincial Children’s Hospital, Children’s Hospital of Fudan University Anhui Hospital, Children’s Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Xiaogang He
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Wei Wang
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Bensheng Qiu
- Department of Electronic Engineering and Information Science, School of Information Science and Technology,
University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Weiping Ding
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Dabing Huang
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| |
Collapse
|
13
|
Zhao B, Dang Z, Li L, Gao J, Wang H, Li M. Development of a simplified and scalable hydrogel-based method for 3D cell culture. Sci Prog 2025; 108:368504251347478. [PMID: 40437988 PMCID: PMC12123143 DOI: 10.1177/00368504251347478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
ObjectiveTo develop a cost-effective and mechanically robust 3D collagen hydrogel system suitable for pressure-based culture, enabling physiologically relevant in vitro modeling of mechanical stress responses in cells.MethodsA rat tail type I collagen-based hydrogel was formulated through optimized component ratios and cast into standard 24-well plates to form uniform gel columns. Endothelial cells was embedded and subjected to 30 mmHg pressure culture for up to 48 h. Gel morphology and fiber architecture were assessed via scanning electron microscopy. Cell viability, proliferation (Ki67 immunostaining), and tube formation ability were evaluated. A custom mechanical compression setup was used to apply and monitor sustained pressure.ResultsThe hydrogel exhibited stable gelation, uniform porosity, and resistance to deformation under mechanical loading. SEM confirmed a consistent nanofiber network, with fiber diameter unaffected by 30 mmHg pressure. After 24-h pressure culture, the gel retained its height and structure. Endothelial cells remained viable but showed reduced proliferation and impaired tube formation under pressure, as indicated by Ki67 staining and angiogenesis assays.ConclusionsThis 3D collagen hydrogel provides a simple, cost-effective, and scalable alternative to complex bioprinting methods, supporting broader application of 3D cell culture in biomedical research.
Collapse
Affiliation(s)
- Bin Zhao
- Department of Pediatric Surgery, The Affiliated Taian City Central Hospital of Qingdao University, Shandong, China
| | - Zhengcheng Dang
- Department of Cardiology, The Affiliated Taian City Central Hospital of Qingdao University, Shandong, China
| | - Lingling Li
- Department of Medical Imaging Center, The Affiliated Taian City Central Hospital of Qingdao University, Shandong, China
| | - Jing Gao
- Department of Stomatology, The Affiliated Taian City Central Hospital of Qingdao University, Shandong, China
| | - Haiyan Wang
- Department of Ultrasound Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, China
| | - Mengzhi Li
- Department of Postdoctoral Workstation, The Affiliated Taian City Central Hospital of Qingdao University, Shandong, China
- Department of Hand, Foot, and Microsurgical Reconstruction, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, China
| |
Collapse
|
14
|
Sousa MGC, Brasino DSK, Krieger M, Dindar DA, Duhen R, Zhang Z, Franca CM, Bertassoni LE. Host-microbe-cancer interactions on-a-chip. Front Bioeng Biotechnol 2025; 13:1505963. [PMID: 40230461 PMCID: PMC11994592 DOI: 10.3389/fbioe.2025.1505963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
The tumor microbiota has emerged as a pivotal contributor to a variety of cancers, impacting disease development, progression, and therapeutic resistance. Due to the complexity of the tumor microenvironment, reproducing the interactions between the microbes, tumor cells, and the immune system remains a great challenge for both in vitro and in vivo studies. To this end, significant progress has been made toward leveraging tumor-on-a-chip model systems to replicate critical hallmarks of the native disease in vitro. These microfluidic platforms offer the ability to mimic essential components of the tumor microenvironment, including controllable fluid flow conditions, manipulable extracellular matrix dynamics, and intricate 3D multi-cellular communication. The primary objective of this review is to discuss recent challenges and advances in engineering host-microbiota and tumor interactions on-a-chip. Ultimately, overcoming these obstacles will help us gain deeper insights into tumor-microbe interactions and enhance avenues for developing more effective cancer therapies.
Collapse
Affiliation(s)
- Mauricio G. C. Sousa
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
| | - Danielle S. K. Brasino
- Department of Microbiology and Molecular Genetics, Robert Larner College of Medicine at the University of Vermont, Burlington, VT, United States
| | - Madeline Krieger
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Duygu A. Dindar
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Rebekka Duhen
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Zhenzhen Zhang
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, United States
| | - Cristiane Miranda Franca
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
| | - Luiz E. Bertassoni
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, United States
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
15
|
Lin YH, Lin CM, Man KM, Hung CC, Hsu HL, Chen Y, Mu HY, Hsiao TH, Huang JH. Real-time and regional analysis of the efficacy of anticancer drugs in a patient-derived intratumoral heterogeneous tumor microenvironment. LAB ON A CHIP 2025; 25:1728-1743. [PMID: 40008895 DOI: 10.1039/d4lc00990h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Preclinical evaluation of anticancer drug efficacy utilizes 2D cell culture systems, tumoroids or experimental animal models, but it suffers from limitations such as inaccurate simulation of tumor microenvironments in living tumors, difficulty in regional analysis, and low throughput. Therefore, in this study, we developed a system named tumor-microenvironment-on-chip (TMoC) comprising a 3D dynamic tumor tissue culture system, which recreated diverse and heterogeneous cellular tumor microenvironments. In addition to the culture with a dynamic circulation, TMoC allowed users to perform real-time regional analysis, independently assessing the drug response from the normoxic area to the hypoxic area in a gradient manner. Through cell composition analysis and gene analysis, we proved that TMoC has a tumor environment with close resemblance to the original tumor environment. By comparing 15 drug testing results with animal experiments, we proved that TMoC is 93% consistent with the response results of animal experiments. In addition, we confirmed that either mouse- or patient-derived tumor cell lines can be cultured and tested in TMoC, indicating its immense potential for all aspects of preclinical drug evaluation.
Collapse
Affiliation(s)
- Ya-Hui Lin
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Chiao-Min Lin
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Kee-Ming Man
- Department of Anesthesiology, China Medical University Hsinchu Hospital, Hsinchu 30272, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
- Department of Medicinal Botanicals and Health Applications, Da Yeh University, Changhua 51591, Taiwan
| | - Chih-Chiang Hung
- Department of Surgery, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Hsin-Ling Hsu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Hsuan-Yu Mu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Tzu-Hung Hsiao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40705, Taiwan.
- Department of Public Health, Fu Jen Catholic University, New Taipei 24205, Taiwan
- Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 40227, Taiwan
| | - Jen-Huang Huang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
16
|
Liu X, Shu Y, Zhu J, Fang H, Su Y, Ma H, Li B, Xu J, Cheng YY, Pan B, Song K. A 3D bioprinted potential colorectal tumor model based on decellularized matrix/gelatin methacryloyl/nanoclay/sodium alginate hydrogel. Int J Biol Macromol 2025; 293:139346. [PMID: 39743054 DOI: 10.1016/j.ijbiomac.2024.139346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/20/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Colorectal cancer (CRC) is now the third most common cancer worldwide. However, the development cycle for anticancer drugs is lengthy and the failure rate is high, highlighting the urgent need for new tumor models for CRC-related research. The decellular matrix (dECM) offers numerous cell adhesion sites, proteoglycan and cytokines. Notably, porcine small intestine is rich in capillaries and lymphatic capillaries, which facilitates nutrient absorption. This study, we utilized dECM, along with methylacryloyl gelatin (GelMA), sodium alginate (SA) and nanoclay (NC) to create a hydrogel scaffold through 3D extrusion bioprinting. Human CRC cells (HCT8) were seeded onto the scaffold and their drug resistance was tested using 5-fluorouracil (5-FU). Our findings indicate that dECM enhances the hydrophilic properties, mechanical strength and biocompatibility of the scaffold. Furthermore, compared to traditional two-dimensional (2D) models, the three-dimensional (3D) scaffold supports the long-term growth of tumor spheres. After 2 days of 5-FU treatment, the cell survival rate reaches 88.06 ± 0.51 %. This suggests that our scaffold provides a promising alternative platform for in vitro research on cancer mechanisms, anti-cancer drug screening and new drug development.
Collapse
Affiliation(s)
- Xinyue Liu
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yan Shu
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Jingjing Zhu
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Huan Fang
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Ya Su
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Hailin Ma
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Bing Li
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Jie Xu
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China.
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, NSW 2007, Australia.
| | - Bo Pan
- Department of Breast Surgery, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou District, Dalian, Liaoning 116023, China.
| | - Kedong Song
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China.
| |
Collapse
|
17
|
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
|
18
|
Handschin C, Shalhoub H, Mazet A, Guyon C, Dusserre N, Boutet-Robinet E, Oliveira H, Guillermet-Guibert J. Biotechnological advances in 3D modeling of cancer initiation. Examples from pancreatic cancer research and beyond. Biofabrication 2025; 17:022008. [PMID: 40018875 DOI: 10.1088/1758-5090/adb51c] [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: 07/19/2024] [Accepted: 02/12/2025] [Indexed: 03/01/2025]
Abstract
In recent years, biofabrication technologies have garnered significant attention within the scientific community for their potential to create advancedin vitrocancer models. While these technologies have been predominantly applied to model advanced stages of cancer, there exists a pressing need to develop pertinent, reproducible, and sensitive 3D models that mimic cancer initiation lesions within their native tissue microenvironment. Such models hold profound relevance for comprehending the intricacies of cancer initiation, to devise novel strategies for early intervention, and/or to conduct sophisticated toxicology assessments of putative carcinogens. Here, we will explain the pivotal factors that must be faithfully recapitulated when constructing these models, with a specific focus on early pancreatic cancer lesions. By synthesizing the current state of research in this field, we will provide insights into recent advances and breakthroughs. Additionally, we will delineate the key technological and biological challenges that necessitate resolution in future endeavors, thereby paving the way for more accurate and insightfulin vitrocancer initiation models.
Collapse
Affiliation(s)
- C Handschin
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - H Shalhoub
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
| | - A Mazet
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - C Guyon
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - N Dusserre
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - E Boutet-Robinet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - H Oliveira
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - J Guillermet-Guibert
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| |
Collapse
|
19
|
Evangelista A, Scocozza F, Conti M, Auricchio F, Conti B, Dorati R, Genta I, Benazzo M, Pisani S. Exploring Mechanical Features of 3D Head and Neck Cancer Models. J Funct Biomater 2025; 16:74. [PMID: 40137353 PMCID: PMC11942903 DOI: 10.3390/jfb16030074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) presents significant challenges in oncology due to its complex biology and poor prognosis. Traditional two-dimensional (2D) cell culture models cannot replicate the intricate tumor microenvironment, limiting their usefulness in studying disease mechanisms and testing therapies. In contrast, three-dimensional (3D) in vitro models provide more realistic platforms that better mimic the architecture, mechanical features, and cellular interactions of HNSCC. This review explores the mechanical properties of 3D in vitro models developed for HNSCC research. It highlights key 3D culture techniques, such as spheroids, organoids, and bioprinted tissues, emphasizing their ability to simulate critical tumor characteristics like hypoxia, drug resistance, and metastasis. Particular attention is given to stiffness, elasticity, and dynamic behavior, highlighting how these models emulate native tumor tissues. By enhancing the physiological relevance of in vitro studies, 3D models offer significant potential to revolutionize HNSCC research and facilitate the development of effective, personalized therapeutic strategies. This review bridges the gap between preclinical and clinical applications by summarizing the mechanical properties of 3D models and providing guidance for developing systems that replicate both biological and mechanical characteristics of tumor tissues, advancing innovation in cancer research and therapy.
Collapse
Affiliation(s)
- Aleksandra Evangelista
- Department of Otorhinolaryngology, Fondazione IRCCS Policlinico San Matteo, Via Golgi 19, 27100 Pavia, Italy; (A.E.); (M.B.)
| | - Franca Scocozza
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy; (M.C.); (F.A.)
| | - Michele Conti
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy; (M.C.); (F.A.)
- 3D and Computer Simulation Laboratory, IRCCS Policlinico San Donato, Piazza Edmondo Malan 2, San Donato Milanese, 20097 Milano, Italy
| | - Ferdinando Auricchio
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy; (M.C.); (F.A.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (B.C.); (R.D.); (I.G.); (S.P.)
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (B.C.); (R.D.); (I.G.); (S.P.)
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (B.C.); (R.D.); (I.G.); (S.P.)
| | - Marco Benazzo
- Department of Otorhinolaryngology, Fondazione IRCCS Policlinico San Matteo, Via Golgi 19, 27100 Pavia, Italy; (A.E.); (M.B.)
| | - Silvia Pisani
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (B.C.); (R.D.); (I.G.); (S.P.)
| |
Collapse
|
20
|
Perucca A, Llonín AG, Benach OM, Hallopeau C, Rivas EI, Linares J, Garrido M, Sallent-Aragay A, Golde T, Colombelli J, Dalaka E, Linacero J, Cazorla M, Galan T, Pastor Viel J, Badenas X, Recort-Bascuas A, Comerma L, Fernandez-Nogueira P, Rovira A, Roca-Cusachs P, Albanell J, Trepat X, Calon A, Labernadie A. Micro Immune Response On-chip (MIRO) models the tumour-stroma interface for immunotherapy testing. Nat Commun 2025; 16:1279. [PMID: 39900918 PMCID: PMC11790944 DOI: 10.1038/s41467-025-56275-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 01/13/2025] [Indexed: 02/05/2025] Open
Abstract
Immunotherapies are beneficial for a considerable proportion of cancer patients, but ineffective in others. In vitro modelling of the complex interactions between cancer cells and their microenvironment could provide a path to understanding immune therapy sensitivity and resistance. Here we develop MIRO, a fully humanised in vitro platform to model the spatial organisation of the tumour/stroma interface and its interaction with immune cells. We find that stromal barriers are associated with immune exclusion and protect cancer cells from antibody-dependent cellular cytotoxicity, elicited by targeted therapy. We demonstrate that IL2-driven immunomodulation increases immune cell velocity and spreading to overcome stromal immunosuppression and restores anti-cancer response in refractory tumours. Collectively, our study underscores the translational value of MIRO as a powerful tool for exploring how the spatial organisation of the tumour microenvironment shapes the immune landscape and influences the responses to immunomodulating therapies.
Collapse
Affiliation(s)
- Alice Perucca
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Andrea Gómez Llonín
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
- Biomaterials, Drug Delivery & Nanotechnology Unit, Centre for Biomedical & Biomaterials Research (CBBR), University of Mauritius, Réduit, Mauritius
| | - Oriol Mañé Benach
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Clement Hallopeau
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Elisa I Rivas
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Jenniffer Linares
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Marta Garrido
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Anna Sallent-Aragay
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Tom Golde
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Julien Colombelli
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Eleni Dalaka
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Judith Linacero
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Marina Cazorla
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Teresa Galan
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Jordi Pastor Viel
- Unitat de Tecnologia Mecànica, Centres Científics i Tecnològics, Universitat de Barcelona, Barcelona, Spain
| | - Xavier Badenas
- Unitat de Tecnologia Mecànica, Centres Científics i Tecnològics, Universitat de Barcelona, Barcelona, Spain
| | - Alba Recort-Bascuas
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Laura Comerma
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
- Pathology Department, Hospital del Mar, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC-ISCIII), Madrid, Spain
| | | | - Ana Rovira
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC-ISCIII), Madrid, Spain
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Unitat de Biofisica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Joan Albanell
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC-ISCIII), Madrid, Spain
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain.
- Unitat de Biofisica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.
- Institucio Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain.
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain.
| | - Alexandre Calon
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain.
| | - Anna Labernadie
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigacion Principe Felipe, Valencia, Spain.
| |
Collapse
|
21
|
Feng QS, Shan XF, Yau V, Cai ZG, Xie S. Facilitation of Tumor Stroma-Targeted Therapy: Model Difficulty and Co-Culture Organoid Method. Pharmaceuticals (Basel) 2025; 18:62. [PMID: 39861125 PMCID: PMC11769033 DOI: 10.3390/ph18010062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/28/2024] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Tumors, as intricate ecosystems, comprise oncocytes and the highly dynamic tumor stroma. Tumor stroma, representing the non-cancerous and non-cellular composition of the tumor microenvironment (TME), plays a crucial role in oncogenesis and progression, through its interactions with biological, chemical, and mechanical signals. This review aims to analyze the challenges of stroma mimicry models, and highlight advanced personalized co-culture approaches for recapitulating tumor stroma using patient-derived tumor organoids (PDTOs). Methods: This review synthesizes findings from recent studies on tumor stroma composition, stromal remodeling, and the spatiotemporal heterogeneities of the TME. It explores popular stroma-related models, co-culture systems integrating PDTOs with stromal elements, and advanced techniques to improve stroma mimicry. Results: Stroma remodeling, driven by stromal cells, highlights the dynamism and heterogeneity of the TME. PDTOs, derived from tumor tissues or cancer-specific stem cells, accurately mimic the tissue-specific and genetic features of primary tumors, making them valuable for drug screening. Co-culture models combining PDTOs with stromal elements effectively recreate the dynamic TME, showing promise in personalized anti-cancer therapy. Advanced co-culture techniques and flexible combinations enhance the precision of tumor-stroma recapitulation. Conclusions: PDTO-based co-culture systems offer a promising platform for stroma mimicry and personalized anti-cancer therapy development. This review underscores the importance of refining these models to advance precision medicine and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Qiu-Shi Feng
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22# Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (Q.-S.F.); (X.-F.S.)
| | - Xiao-Feng Shan
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22# Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (Q.-S.F.); (X.-F.S.)
| | - Vicky Yau
- Division of Oral and Maxillofacial Surgery, Columbia Irving Medical Center, New York City, NY 10027, USA;
| | - Zhi-Gang Cai
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22# Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (Q.-S.F.); (X.-F.S.)
| | - Shang Xie
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22# Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (Q.-S.F.); (X.-F.S.)
| |
Collapse
|
22
|
Alvarez MM, Cantoral-Sánchez A, Trujillo-de Santiago G. Chaotic (bio)printing in the context of drug delivery systems. Adv Drug Deliv Rev 2025; 216:115475. [PMID: 39561907 DOI: 10.1016/j.addr.2024.115475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/26/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024]
Abstract
Chaotic (bio)printing, an innovative fabrication technique that uses chaotic flows to create highly ordered microstructures within materials, may be transformative for drug delivery systems. This review explores the principles underlying chaotic flows and their application in fabricating complex, multi-material constructs designed for advanced drug delivery and controlled release. Chaotic printing enables the precise layering of different active ingredients-a feature that may greatly facilitate the development of polypills with customizable release profiles. Recently, chaos-assisted fabrication has been extended to produce micro-architected hydrogel spheres in a high-throughput manner, potentially enhancing the versatility and efficiency of drug delivery methods. In addition, chaotic bioprinting enables the creation of evolved tissue models that more accurately emulate physiological systems, providing a more relevant platform for drug testing. This review also highlights the unique advantages of chaotic printing, including the ability to fabricate tissues with organized porosity and pre-vascularized structures, addressing critical challenges in tissue engineering. Despite its promising capabilities, challenges remain, particularly in expanding the range of materials compatible with chaotic printing. Continued research and development in this area are essential to fully realize the potential of chaotic (bio)printing in advancing drug delivery, paving the way for the next generation of smart drug delivery systems and functional tissue models for drug testing.
Collapse
Affiliation(s)
- Mario Moisés Alvarez
- Centro de Biotecnología-FEMSA, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL 64849, Mexico; Departamento de Ingeniería Mecatrónica y Eléctrica, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL 64849, Mexico; Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL 64849, Mexico.
| | - Ariel Cantoral-Sánchez
- Centro de Biotecnología-FEMSA, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL 64849, Mexico; Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL 64849, Mexico
| | - Grissel Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL 64849, Mexico; Departamento de Ingeniería Mecatrónica y Eléctrica, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL 64849, Mexico; Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL 64849, Mexico.
| |
Collapse
|
23
|
Li Y, Orange JS. A Thermo-responsive collapse system for controlling heterogeneous cell localization, ratio and interaction for three-dimensional solid tumor modeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.26.630018. [PMID: 39764015 PMCID: PMC11703237 DOI: 10.1101/2024.12.26.630018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Cancer immunotherapy using engineered cytotoxic effector cells has demonstrated significant potential. The limited spatial complexity of existing in vitro models, however, poses a challenge to mechanistic studies attempting to approve existing approaches of effector cell-mediated cytotoxicity within a three-dimensional, solid tumor-like environment. To gain additional experimental control, we developed an approach for constructing three-dimensional (3D) culture models using smart polymers that form temperature responsive hydrogels. By embedding cells in these hydrogels, we constructed 3D models to organize multiple cell populations at specified ratios on-demand and gently position them by exploiting the hydrogel phase transition. These systems were amenable to imaging at low- and high-resolution to evaluate cell-to-cell interactions, as well as to dissociation to allow for single cell analyses. We have called this approach "thermal collapse of strata" (TheCOS) and demonstrated its use in creating complex cell assemblies on demand in both layers and spheroids. As an application, we utilized TheCOS to evaluate the impact of directionality of degranulation of natural killer (NK) cell lytic granules. Blocking lytic granule convergence and polarization by inhibiting dynein has been shown to induce bystander killing in single cell suspensions. Using TheCOS we showed that lytic granule dispersion induced by dynein inhibition can be sustained in 3D and results in a multi-directional killing including that of non-triggering bystander cells. By imaging TheCOS experiments, we were able to map a "kill zone" associated with multi-directional degranulation in simulated solid tumor environments. TheCOS should allow for the testing of approaches to alter the mechanics of cytotoxicity as well as to generate a wide-array of human tumor microenvironments to assist in the acceleration of tumor immunotherapy.
Collapse
Affiliation(s)
- Yu Li
- Columbia University Vagelos College of Physicians and Surgeons
| | - Jordan S Orange
- Columbia University Vagelos College of Physicians and Surgeons
| |
Collapse
|
24
|
Nunes M, Ricardo S. Ivermectin Strengthens Paclitaxel Effectiveness in High-Grade Serous Carcinoma in 3D Cell Cultures. Pharmaceuticals (Basel) 2024; 18:14. [PMID: 39861076 PMCID: PMC11769219 DOI: 10.3390/ph18010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Chemoresistance is a major obstacle in high-grade serous carcinoma (HGSC) treatment. Although many patients initially respond to chemotherapy, the majority of them relapse due to Carboplatin and Paclitaxel resistance. Drug repurposing has surfaced as a potentially effective strategy that works synergically with standard chemotherapy to bypass chemoresistance. In a prior study, using 2D cultures and two HGSC chemoresistant cell lines, it was demonstrated that combining Carboplatin or Paclitaxel with Pitavastatin or Ivermectin resulted in the most notable synergy. Acknowledging that 2D culture systems are limited in reflecting the tumor architecture, 3D cultures were generated to provide insights on treatment efficacy tests in more complex models. OBJECTIVES We aimed to investigate whether combining Carboplatin or Paclitaxel with Pitavastatin or Ivermectin offers therapeutic benefits in a Cultrex-based 3D model. METHODS Here, the cytotoxicity of Carboplatin and Paclitaxel, both alone and in combination with Pitavastatin or Ivermectin, were analyzed on two chemoresistant tumor cell lines, OVCAR8 and OVCAR8 PTX R C, in 3D cultures. Cellular viability was assessed using CellTiter-Glo® Luminescent assays. Also, it explored synergistic interactions using zero interaction potency, Loewe, Bliss independence, and High-single agent reference models. RESULTS Our research indicates combining chemotherapeutic drugs with Pitavastatin or Ivermectin yields significantly more cytotoxic effects than chemotherapy alone. For all the combinations tested, at least one model indicated an additive effect; however, only the combination of Paclitaxel and Ivermectin consistently demonstrated an additive effect across all chemoresistant cell lines cultured in 3D models, as well as in all four synergy reference models used to assess drug interactions. CONCLUSIONS Combining Paclitaxel with Ivermectin has the highest cytotoxic and the strongest additive effect for both chemoresistant cell lines compared to Paclitaxel alone.
Collapse
Affiliation(s)
- Mariana Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135 Porto, Portugal;
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135 Porto, Portugal;
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences (IUCS), University Polytechnic Higher Education Cooperative (CESPU), CRL, 4585-116 Gandra, Portugal
- Applied Molecular Biosciences Unit (UCIBIO), Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
- Oral Pathology and Rehabilitation Research Unit (UNIPRO), Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| |
Collapse
|
25
|
Luo Y, Gadd ME, Qie Y, Otamendi-Lopez A, Sanchez-Garavito JE, Brooks MM, Ulloa Navas MJ, Hundal T, Li S, Jones VK, Lou Y, Patel T, Dronca R, Kharfan-Dabaja MA, Dong H, Quinones-Hinojosa A, Qin H. Solid cancer-directed CAR T cell therapy that attacks both tumor and immunosuppressive cells via targeting PD-L1. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200891. [PMID: 39498357 PMCID: PMC11532918 DOI: 10.1016/j.omton.2024.200891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/31/2024] [Accepted: 10/03/2024] [Indexed: 11/07/2024]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has encountered limited success in solid tumors. The lack of dependable antigens and the immunosuppressive tumor microenvironment (TME) are major challenges. Within the TME, tumor cells along with immunosuppressive cells employ an immune-evasion mechanism that upregulates programmed death ligand 1 (PD-L1) to deactivate effector T cells; this makes PD-L1 a reliable, universal target for solid tumors. We developed a novel PD-L1 CAR (MC9999) using our humanized anti-PD-L1 monoclonal antibody, designed to simultaneously target tumor and immunosuppressive cells. The antigen-specific antitumor effects of MC9999 CAR T cells were observed consistently across four solid tumor models: breast cancer, lung cancer, melanoma, and glioblastoma multiforme (GBM). Notably, intravenous administration of MC9999 CAR T cells eradicated intracranially established LN229 GBM tumors, suggesting penetration of the blood-brain barrier. The proof-of-concept data demonstrate the cytolytic effect of MC9999 CAR T cells against immunosuppressive cells, including microglia HMC3 cells and M2 macrophages. Furthermore, MC9999 CAR T cells elicited cytotoxicity against primary tumor-associated macrophages within GBM tumors. The concept of targeting both tumor and immunosuppressive cells with MC9999 was further validated using CAR T cells derived from cancer patients. These findings establish MC9999 as a foundation for the development of effective CAR T cell therapies against solid tumors.
Collapse
Affiliation(s)
- Yan Luo
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Martha E. Gadd
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
| | - Yaqing Qie
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Mieu M. Brooks
- The Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA
| | | | - Tanya Hundal
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
| | - Shuhua Li
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
| | | | - Yanyan Lou
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Tushar Patel
- Hepatology & Liver Transplantation, Mayo Clinic, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Roxana Dronca
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Mohamed A. Kharfan-Dabaja
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
- Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | - Haidong Dong
- Department of Urology, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Alfredo Quinones-Hinojosa
- The Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Hong Qin
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
26
|
Muhammad FA, Altalbawy FMA, Mandaliya V, Saraswat SK, Rekha MM, Aulakh D, Chahar M, Mahdi MS, Jaber MA, Alhadrawi M. Targeting breast tumor extracellular matrix and stroma utilizing nanoparticles. Clin Transl Oncol 2024:10.1007/s12094-024-03793-x. [PMID: 39692807 DOI: 10.1007/s12094-024-03793-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024]
Abstract
Breast cancer is a complicated malignancy and is known as the most common cancer in women. Considerable experiments have been devoted to explore the basic impacts of the tumor stroma, particularly the extracellular matrix (ECM) and stromal components, on tumor growth and resistance to treatment. ECM is made up of an intricate system of proteins, glycosaminoglycans, and proteoglycans, and maintains structural support and controls key signaling pathways involved in breast tumors. ECM can block different drugs such as chemotherapy and immunotherapy drugs from entering the tumor stroma. Furthermore, the stromal elements, such as cancer-associated fibroblasts (CAFs), immune cells, and blood vessels, have crucial impacts on tumor development and therapeutic resistance. Recently, promising outcomes have been achieved in using nanotechnology for delivering drugs to tumor stroma and crossing ECM in breast malignancies. Nanoparticles have various benefits for targeting the breast tumor stroma, such as improved permeability and retention, extended circulation time, and the ability to actively target the area. This review covers the latest developments in nanoparticle therapies that focus on breast tumor ECM and stroma. We will explore different approaches using nanoparticles to target the delivery of anticancer drugs like chemotherapy, small molecule drugs, various antitumor products, and other specific synthetic therapeutic agents to the breast tumor stroma. Furthermore, we will investigate the utilization of nanoparticles in altering the stromal elements, such as reprogramming CAFs and immune cells, and also remodeling ECM.
Collapse
Affiliation(s)
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, 12613, Egypt.
| | - Viralkumar Mandaliya
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | | | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Damanjeet Aulakh
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology Chitkara University, Rajpura, Punjab, 140401, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering and Technology, NIMS University Rajasthan, Jaipur, India
| | | | | | - Merwa Alhadrawi
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
27
|
Ielpo S, Barberini F, Dabbagh Moghaddam F, Pesce S, Cencioni C, Spallotta F, De Ninno A, Businaro L, Marcenaro E, Bei R, Cifaldi L, Barillari G, Melaiu O. Crosstalk and communication of cancer-associated fibroblasts with natural killer and dendritic cells: New frontiers and unveiled opportunities for cancer immunotherapy. Cancer Treat Rev 2024; 131:102843. [PMID: 39442289 DOI: 10.1016/j.ctrv.2024.102843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
Natural killer (NK) cells and dendritic cells (DCs) are critical mediators of anti-cancer immune responses. In addition to their individual roles, NK cells and DCs are involved in intercellular crosstalk which is essential for the initiation and coordination of adaptive immunity against cancer. However, NK cell and DC activity is often compromised in the tumor microenvironment (TME). Recently, much attention has been paid to one of the major components of the TME, the cancer-associated fibroblasts (CAFs), which not only contribute to extracellular matrix (ECM) deposition and tumor progression but also suppress immune cell functions. It is now well established that CAFs support T cell exclusion from tumor nests and regulate their cytotoxic activity. In contrast, little is currently known about their interaction with NK cells, and DCs. In this review, we describe the interaction of CAFs with NK cells and DCs, by secreting and expressing various mediators in the TME of adult solid tumors. We also provide a detailed overview of ongoing clinical studies evaluating the targeting of stromal factors alone or in combination with immunotherapy based on immune checkpoint inhibitors. Finally, we discuss currently available strategies for the selective depletion of detrimental CAFs and for a better understanding of their interaction with NK cells and DCs.
Collapse
Affiliation(s)
- Simone Ielpo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Barberini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Farnaz Dabbagh Moghaddam
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, Rome, Italy
| | - Silvia Pesce
- Department of Experimental Medicine and Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Chiara Cencioni
- Institute for Systems Analysis and Computer Science "A. Ruberti", National Research Council (IASI-CNR), Rome, Italy
| | - Francesco Spallotta
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University, 00185, Rome, Italy; Pasteur Institute Italy-Fondazione Cenci Bolognetti, Italy
| | - Adele De Ninno
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, Rome, Italy
| | - Luca Businaro
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, Rome, Italy
| | - Emanuela Marcenaro
- Department of Experimental Medicine and Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Loredana Cifaldi
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
28
|
Spiteri C, Caprettini V, Wang Y, Dominguez‐Gil S, Kaasalainen M, Wang C, Martella DA, McLennan S, Vashisth P, Gary‐Bobo M, Nguyen C, Bergholt M, Durand J, Cunin F, Chiappini C. Spatially-Resolved Organoid Transfection by Porous Silicon-Mediated Optoporation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407650. [PMID: 39420545 PMCID: PMC11619231 DOI: 10.1002/adma.202407650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/27/2024] [Indexed: 10/19/2024]
Abstract
Engineering the spatial organisation of organotypic cultures is pivotal for refining tissue models that are useful for gaining deeper insights into complex, non-cell autonomous processes. These advanced models are key to improving the understanding of fundamental biological mechanisms and therapeutic strategies. Controlling gene regulation through spatially-resolved delivery of nucleic acids provides an attractive approach to produce such tissue models. An emerging strategy for spatially-resolved transfection uses photosensitizing nanoparticles coupled with laser pulses to optoporate cells in culture and locally mediate gene delivery. However, localized optoporation in 3D systems remains challenging. Here we propose a solution to this longstanding hurdle, demonstrating that porous silicon nanoparticles are a safe and bioresorbable photosensitising nanomaterial capable of spatially-resolved transfection of mRNA in MCF-7 organoids by near-infrared two-photon optoporation. Functionalization with an azobenzene-lysine photo-switchable moiety enhances the transfection efficiency of the nanoparticles up to 84% in a 2D cell system. Moreover, the nanoparticles enable spatially selective mRNA transfection to MCF-7 spheroids, demonstrating targeted gene delivery in complex 3D cellular environments. The approach for spatially-resolved 3D optoporation offers a way forward for the design of tailored spheroids and organoids by spatially selective nucleic acids delivery.
Collapse
Affiliation(s)
- Chantelle Spiteri
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | - Valeria Caprettini
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | - Yikai Wang
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | | | - Martti Kaasalainen
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | - Cong Wang
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | - Davide Alessandro Martella
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | - Samuel McLennan
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| | - Priya Vashisth
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
| | | | | | - Mads Bergholt
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
| | | | | | - Ciro Chiappini
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUnited Kingdom
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUnited Kingdom
| |
Collapse
|
29
|
Da Silva André G, Labouesse C. Mechanobiology of 3D cell confinement and extracellular crowding. Biophys Rev 2024; 16:833-849. [PMID: 39830117 PMCID: PMC11735831 DOI: 10.1007/s12551-024-01244-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 09/30/2024] [Indexed: 01/22/2025] Open
Abstract
Cells and tissues are often under some level of confinement, imposed by the microenvironment and neighboring cells, meaning that there are limitations to cell size, volume changes, and fluid exchanges. 3D cell culture, increasingly used for both single cells and organoids, inherently impose levels of confinement absent in 2D systems. It is thus key to understand how different levels of confinement influences cell survival, cell function, and cell fate. It is well known that the mechanical properties of the microenvironment, such as stiffness and stress relaxation, are important in activating mechanosensitive pathways, and these are responsive to confinement conditions. In this review, we look at how low, intermediate, and high levels of confinement modulate the activation of known mechanobiology pathways, in single cells, organoids, and tumor spheroids, with a specific focus on 3D confinement in microwells, elastic, or viscoelastic scaffolds. In addition, a confining microenvironment can drastically limit cellular communication in both healthy and diseased tissues, due to extracellular crowding. We discuss potential implications of extracellular crowding on molecular transport, extracellular matrix deposition, and fluid transport. Understanding how cells sense and respond to various levels of confinement should inform the design of 3D engineered matrices that recapitulate the physical properties of tissues.
Collapse
Affiliation(s)
- Gabriela Da Silva André
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Céline Labouesse
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
30
|
Liu X, Sun M, Zhang F, Zhang J, Xuanyuan T, Liu W. A heterotypic tumor-on-a-chip platform for user-friendly combinatorial chemotherapeutic testing. Anal Chim Acta 2024; 1330:343278. [PMID: 39489960 DOI: 10.1016/j.aca.2024.343278] [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: 07/22/2024] [Revised: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Three-dimensional (3D) tumor microdevices are promising platform for biomimetic antitumor prediction and high-throughput chemotherapeutic screening and play crucial roles in the exploration of cancer-associated pharmaceutics and therapeutics. Traditional cell manipulation tools (e.g., non-adhesive surfaces and hanging drops) and recent microengineered systems (e.g., microfluidic chips and micropatterned array chips) have progressed in terms of microscale control, substantial tumor production, programmable drug combinations, and throughput analysis. However, establishing a facile 3D tumor microdevice to construct heterotypic tumor-microenvironmental profiles and for throughput, implementable, multi-instrument-compatible analysis of chemotherapies to advance consumer-grade tumor modelling tools is still being explored. RESULTS In this study, we present a facilely operated tumor-on-a-chip platform for massive production of heterotypic 3D tumors and diverse investigations of combinatorial chemotherapy screening. Large quantity of heterotypic tumor generation with high geometric controllability (size difference: 19.6 μm) and operational repeatability (n = 10) was achieved using simple-to-fabricate micropatterned chips. Multiple characteristics of solid tumors, including phenotypic gradients (viability and proliferation) and heterogeneous cellular compositions (multi-cell participation and stroma composition), were reproduced in heterotypic tumors, being more biomimetic than homotypic tumors. We completed the user-friendly analytical evaluation of individual and combinatorial drug therapies, and demonstrated the high applicability of the platform in biomimetic tumor-related large-scale manipulation and on-chip analysis, as well as its high compatibility for off-chip detection. The entire operative process during tumor production and chemotherapy only requires the routine and easy-to-master pipetting manipulation. SIGNIFICANCE The establishment of a biomimetic and easy-to-use 3D tumor platform and the large-scale screening-like evaluation of combinatorial chemotherapies based on the usage of the micropatterned chip was achieved in a user-friendly manner. This advancement has significant application potential in the fields of oncology, drug discovery, and tissue engineering, and is expected to be valuable for developing accessible and generalizable tumor-on-a-chip microsystems for exploring cancer therapies.
Collapse
Affiliation(s)
- Xufang Liu
- Departments of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Meilin Sun
- Departments of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Fen Zhang
- Departments of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China; Department of Bioengineering, School of Food and Bioengineering, Xuzhou University of Technology, Xuzhou, 221018, China.
| | - Jinwei Zhang
- Departments of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Tingting Xuanyuan
- Departments of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Wenming Liu
- Departments of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China.
| |
Collapse
|
31
|
Pape J, Cheema U, Tocci P, Sestito R, Masi I, Loizidou M, Bagnato A, Rosanò L. Endothelin-1 receptor blockade impairs invasion patterns in engineered 3D high-grade serous ovarian cancer tumouroids. Clin Sci (Lond) 2024; 138:1441-1450. [PMID: 39503511 PMCID: PMC11570180 DOI: 10.1042/cs20240371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
High-grade serous ovarian cancer (HG-SOC), accounting for 70-80% of ovarian cancer deaths, is characterized by a widespread and rapid metastatic nature, influenced by diverse cell types, cell-cell interactions, and acellular components of the tumour microenvironment (TME). Within this tumour type, autocrine and paracrine activation of the endothelin-1 receptors (ET-1R), expressed in tumour cells and stromal elements, drives metastatic progression. The lack of three-dimensional models that faithfully recapitulate the unique HG-SOC TME has been the bottleneck in performing drug screening for personalized medicine. Herein, we developed HG-SOC tumouroids by engineering a dense central artificial cancer mass (ACM) containing HG-SOC cells, nested within a compressed hydrogel recapitulating the stromal compartment comprising type I collagen, laminin, fibronectin, and stromal cells (fibroblasts and endothelial cells). ET-1-stimulated HG-SOC cells in the tumouroids showed an altered migration pattern and formed cellular aggregates, mimicking micrometastases that invaded the stroma. Compared with control cells, ET-1-stimulated tumouroids showed a higher number of invasive bodies, which were reduced by treatment with the dual ET-1 receptor (ET-1R) antagonist macitentan. In addition, ET-1 increased the size of the invading aggregates compared with control cells. This study establishes an experimental 3D multicellular model eligible for mechanical research, investigating the impact of matrix stiffness and TME interactions, which will aid drug screening to guide therapeutic decisions in HG-SOC patients.
Collapse
Affiliation(s)
- Judith Pape
- UCL Division of Surgery and Interventional Science, London, U.K
| | - Umber Cheema
- UCL Division of Surgery and Interventional Science, London, U.K
| | - Piera Tocci
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Rosanna Sestito
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Ilenia Masi
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), Rome, 00185, Italy
| | | | - Anna Bagnato
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Rosanò
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), Rome, 00185, Italy
| |
Collapse
|
32
|
Rakhmatullina AR, Zolotykh MA, Filina YV, Valiullina AK, Zmievskaya EA, Gafurbaeva DU, Sagdeeva AR, Bulatov ER, Rizvanov AA, Miftakhova RR. Multicellular Cancer-Stroma Spheres (CSS) for In Vitro Assessment of CAR-T Cell-Associated Toxicity. Cells 2024; 13:1892. [PMID: 39594640 PMCID: PMC11593285 DOI: 10.3390/cells13221892] [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: 10/09/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
CAR-T therapy has revolutionized the field of oncology, offering a promising treatment option for cancer patients. However, the significant morbidity associated with therapy-related toxicity presents a major challenge to its widespread use. Despite extensive research into the underlying mechanisms of CAR-T therapy-related toxicity, there are still many unknowns. Furthermore, the lack of adequate in vitro models for assessing immunotoxicity and neurotoxicity further complicates the development of safer cellular therapies. Previously in our laboratory, we developed cancer-stroma spheres (CSS) composed of prostate adenocarcinoma PC3 cells and mesenchymal stem cells (MSC). Herein we present evidence that multicellular CSS could serve as a valuable in vitro model for toxicity studies related to CAR-T therapy. CSS containing CD19-overexpressing PC3M cells exhibited increased secretion of CAR-T cell toxicity-associated IL-8, MCP-1, and IP-10 in the presence of anti-CD19 CAR-T cells, compared to spheres derived from single cell types.
Collapse
Affiliation(s)
- Aigul R. Rakhmatullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Mariya A. Zolotykh
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Yuliya V. Filina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Aigul Kh. Valiullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Ekaterina A. Zmievskaya
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Dina U. Gafurbaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Aisylu R. Sagdeeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Emil R. Bulatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Division of Medical and Biological Sciences, Academy of Sciences of the Republic of Tatarstan, 420111 Kazan, Russia
| | - Regina R. Miftakhova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
33
|
Barros da Silva P, Zhao X, Bidarra SJ, Nascimento DS, LaLone V, Lourenço BN, Paredes J, Stevens MM, Barrias CC. Tunable Hybrid Hydrogels of Alginate and Cell-Derived dECM to Study the Impact of Matrix Alterations on Epithelial-to-Mesenchymal Transition. Adv Healthc Mater 2024; 13:e2401032. [PMID: 39246099 PMCID: PMC11582509 DOI: 10.1002/adhm.202401032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/27/2024] [Indexed: 09/10/2024]
Abstract
Epithelial-to-mesenchymal transition (EMT) is crucial for tumor progression, being linked to alterations in the extracellular matrix (ECM). Understanding the ECM's role in EMT can uncover new therapeutic targets, yet replicating these interactions in vitro remains challenging. It is shown that hybrid hydrogels of alginate (ALG) and cell-derived decellularized ECM (dECM), with independently tunable composition and stiffness, are useful 3D-models to explore the impact of the breast tumor matrix on EMT. Soft RGD-ALG hydrogels (200 Pa), used as neutral bulk material, supported mammary epithelial cells morphogenesis without spontaneous EMT, allowing to define the gene, protein, and biochemical profiles of cells at different TGFβ1-induced EMT states. To mimic the breast tumor composition, dECM from TGFβ1-activated fibroblasts (adECM) are generated, which shows upregulation of tumor-associated proteins compared to ndECM from normal fibroblasts. Using hybrid adECM-ALG hydrogels, it is shown that the presence of adECM induces partial EMT in normal epithelial cells, and amplifes TGF-β1 effects compared to ALG and ndECM-ALG. Increasing the hydrogel stiffness to tumor-like levels (2.5 kPa) have a synergistic effect, promoting a more evident EMT. These findings shed light on the complex interplay between matrix composition and stiffness in EMT, underscoring the utility of dECM-ALG hydrogels as a valuable in vitro platform for cancer research.
Collapse
Affiliation(s)
- P Barros da Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Porto, 4200-135, Portugal
| | - Xiaoyu Zhao
- Department of Bioengineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Sílvia J Bidarra
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
| | - Diana S Nascimento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4200-135, Portugal
| | - Vernon LaLone
- Department of Bioengineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Department of Materials, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Bianca N Lourenço
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Porto, 4200-135, Portugal
| | - Joana Paredes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- FMUP - Faculdade de Medicina da Universidade do Porto, Porto, 4200-319, Portugal
- IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, 4200-135, Portugal
| | - Molly M Stevens
- Department of Bioengineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
- Department of Materials, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - C C Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4200-135, Portugal
| |
Collapse
|
34
|
Wu C, Zhai Y, Ji J, Yang X, Ye L, Lu G, Shi X, Zhai G. Advances in tumor stroma-based targeted delivery. Int J Pharm 2024; 664:124580. [PMID: 39142464 DOI: 10.1016/j.ijpharm.2024.124580] [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: 05/13/2024] [Revised: 08/06/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
The tumor stroma plays a crucial role in tumor progression, and the interactions between the extracellular matrix, tumor cells, and stromal cells collectively influence tumor progression and the efficacy of therapeutic agents. Currently, utilizing components of the tumor stroma for drug delivery is a noteworthy strategy. A number of targeted drug delivery systems designed based on tumor stromal components are entering clinical trials. Therefore, this paper provides a thorough examination of the function of tumor stroma in the advancement of targeted drug delivery systems. One approach is to use tumor stromal components for targeted drug delivery, which includes certain stromal components possessing inherent targeting capabilities like HA, laminin, along with targeting stromal cells homologously. Another method entails directly focusing on tumor stromal components to reshape the tumor stroma and facilitate drug delivery. These drug delivery systems exhibit great potential in more effective cancer therapy strategies, such as precise targeting, enhanced penetration, improved safety profile, and biocompatibility. Ultimately, the deployment of these drug delivery systems can deepen our comprehension of tumor stroma and the advanced development of corresponding drug delivery systems.
Collapse
Affiliation(s)
- Chunyan Wu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
35
|
Seo J, Ha G, Lee G, Nasiri R, Lee J. Modeling tumor-immune interactions using hybrid spheroids and microfluidic platforms for studying tumor-associated macrophage polarization in melanoma. Acta Biomater 2024:S1742-7061(24)00629-9. [PMID: 39461691 DOI: 10.1016/j.actbio.2024.10.036] [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: 07/16/2024] [Revised: 10/19/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
Tumor-associated macrophages (TAMs), as key components of tumor microenvironment (TME), exhibit phenotypic plasticity in response to environmental cues, causing polarization into either pro-inflammatory M1 phenotypes or immunosuppressive M2 phenotypes. Although TAM has been widely studied for its crucial involvement in the initiation, progression, metastasis, and immune regulation of cancer cells, there have been limited attempts to understand how the metastatic potentials of cancer cells influence TAM polarization within TME. Here, we developed a miniaturized TME model using a 3D hybrid system composed of murine melanoma cells and macrophages, aiming to investigate interactions between cancer cells exhibiting various metastatic potentials and macrophages within TME. The increase in spheroid size within this model was associated with a reduction in cancer cell viability. Examining macrophage surface marker expression and cytokine secretion indicated the development of diverse TMEs influenced by both spheroid size and the metastatic potential of cancer cells. Furthermore, a high-throughput microfluidic platform equipped with trapping systems and hybrid spheroids was employed to simulate the tumor-immune system of complex TMEs and for comparative analysis with traditional 3D culture models. This study provides insight into TAM polarization in melanoma with different heterogeneities by modeling cancer-immune systems, which can be potentially employed for immune-oncology research, drug screening, and personalized therapy. STATEMENT OF SIGNIFICANCE: This study presents the development of a 3D hybrid spheroid system designed to model tumor-immune interactions, providing a detailed analysis of how melanoma cell metastatic potential influences tumor-associated macrophage (TAM) polarization. By utilizing a microfluidic platform, we are able to replicate and investigate the complex tumor-immune system of the tumor microenvironments (TMEs) under continuous flow conditions. Our model holds significant potential for high-throughput drug screening and personalized medicine applications, offering a versatile tool for advancing cancer research and treatment strategies.
Collapse
Affiliation(s)
- Junki Seo
- Division of Interdisciplinary Bioscience & Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Giheon Ha
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Geonho Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Rohollah Nasiri
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, United States
| | - Junmin Lee
- Division of Interdisciplinary Bioscience & Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea; Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Incheon 21983, Republic of Korea.
| |
Collapse
|
36
|
Jiang Y, Hao M, Chen S, Xie Y, Liu K. Exploring the impact of microfluidic chip structure on the efficacy of three-dimensional tumor microspheres cultivation. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:7005-7014. [PMID: 39282915 DOI: 10.1039/d4ay01343c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Three-dimensional (3D) tumor microspheres can simulate the interaction and growth dynamics of tumor cells, and have been used as a new in vitro model for drug screening and tumor biology related research. The scaffold-free culture of 3D tumor microspheres on microfluidic chips has many advantages, including low cost, high throughput, convenience and flexibility. However, it is still unclear how various factors, such as chip structure, influence the culture effect of tumor microspheres. The lack of standardized evaluation and characterization of the culture effect hinders the further optimization and development of chip function. This study presents numerical simulations of multiple parts or processes of the proposed 3D culture chips with two different structural parameters based on computational fluid dynamics (CFD) methods. An evaluation system for tumor microspheres was established. The prediction of the CFD simulation was consistent with the culture results of the chips, reflecting the important role of the structural parameters of the microtrap in the formation of uniform tumor microspheres. Furthermore, the velocity of cell suspension also had a significant impact on the retention of tumor cells. Additionally, the drug screening results of tumor microspheres indicated that tumor microspheres exhibit greater drug resistance, which may be attributed to their size. These results offer valuable insights into the factors that influence the characteristics of tumor microspheres. This research provides a reference and direction for the optimal design and functional evaluation of scaffold-free 3D culture chips, and holds promise for promoting the development of novel drug research platforms.
Collapse
Affiliation(s)
- Yue Jiang
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China.
| | - Ming Hao
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China.
| | - Shulei Chen
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China.
| | - Yuanhua Xie
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China.
| | - Kun Liu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China.
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, China
| |
Collapse
|
37
|
Poddar MS, Chu YD, Pendharkar G, Liu CH, Yeh CT. Exploring cancer-associated fibroblast-induced resistance to tyrosine kinase inhibitors in hepatoma cells using a liver-on-a-chip model. LAB ON A CHIP 2024; 24:5043-5054. [PMID: 39356081 DOI: 10.1039/d4lc00624k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Liver cancer is a significant global contributor to cancer-related mortality. Despite available targeted therapies, resistance to tyrosine kinase inhibitors (TKIs) like sorafenib and lenvatinib poses a formidable challenge. The tumor microenvironment (TME), inhabited by cancer-associated fibroblasts (CAFs), profoundly influences this resistance. To uncover the mechanisms, a 3D microfluidic chip replicating liver architecture was fabricated to probe the intricate mechanisms of TKI resistance. The chip design mirrors the hexagonal structure of liver lobules, situating liver cancer cells at the core, encircled by fibroblasts, with rigorous assessments confirming biocompatibility and consistent cell growth. After determining the IC50 values of sorafenib and lenvatinib in 2D co-culture, a transwell setup revealed drug resistance development in co-cultured cells. Within the 3D microfluidic chip, live/dead assays highlighted elevated viability under drug exposure, emphasizing fibroblast-driven drug resistance. The study identifies AHSG and CLEC3B as potential mediators of drug resistance in co-culture, significantly upregulated in the co-cultured medium. Functional tests confirmed their roles, as introducing recombinant AHSG and CLEC3B enhanced liver cancer cell resistance to sorafenib and lenvatinib in both 2D and 3D scenarios. In conclusion, by replicating the complex TME using microfluidic technology, this study sheds light on the roles of AHSG and CLEC3B as well as possible approaches for improving the effectiveness of liver cancer treatment.
Collapse
Affiliation(s)
- Madhu Shree Poddar
- Institute of Nanoengineering and Microsystems, National Tsing Hua University, Hsinchu, 30044, Taiwan, R.O.C..
| | - Yu-De Chu
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan, R.O.C..
| | - Gaurav Pendharkar
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 30044, Taiwan, R.O.C
| | - Cheng-Hsien Liu
- Institute of Nanoengineering and Microsystems, National Tsing Hua University, Hsinchu, 30044, Taiwan, R.O.C..
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 30044, Taiwan, R.O.C
- College of Semiconductor Research, National Tsing Hua University, Hsinchu 30044, Taiwan, R.O.C
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan, R.O.C..
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan, R.O.C
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| |
Collapse
|
38
|
Barros da Silva P, Oliveira RJA, Araújo M, Caires HR, Bidarra SJ, Barrias CC. An integrative alginate-based 3D in vitro model to explore epithelial-stromal cell dynamics in the breast tumor microenvironment. Carbohydr Polym 2024; 342:122363. [PMID: 39048221 DOI: 10.1016/j.carbpol.2024.122363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/10/2024] [Accepted: 06/03/2024] [Indexed: 07/27/2024]
Abstract
The tumor microenvironment (TME) orchestrates cellular and extracellular matrix (ECM) interactions, playing a key role in tumorigenesis, tumor growth, and metastization. Investigating the interplay between stromal-epithelial cells within the TME is paramount for understanding cancer mechanisms but demands reliable biological models. 3D-models have emerged as powerful in vitro tools, but many fall short in replicating cell-cell/cell-matrix interactions. This study introduces a novel hybrid 3D-model of the breast TME, combining epithelial cells, cancer-associated fibroblasts (CAFs), and their ECM. To build the stromal compartment, porous 3D-printed alginate scaffolds were seeded with CAFs, which proliferated and produced ECM. The pores were infused with oxidized peptide-modified alginate hydrogel laden with MCF10A cells, forming the parenchymal compartment. The hybrid system supported epithelial morphogenesis into acini surrounded by fibroblasts and ECM, and could be readily solubilized to recover cells, their matrix, and sequestered soluble factors. Proteome profiling of the retrieved ECM showed upregulation of proteins associated with matrix assembly/remodeling, epithelial-to-mesenchymal transition (EMT), and cancer. The TME-like microenvironment induced a partial EMT in MCF10A cells, generating a hybrid population with epithelial and mesenchymal features, characteristic of aggressive phenotypes. Our model provided new insights into epithelial-stromal interactions within the TME, offering a valuable tool for cancer research in a physiologically-relevant 3D setting.
Collapse
Affiliation(s)
- P Barros da Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 5 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | - R J A Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 5 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - M Araújo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 5 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - H R Caires
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 5 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - S J Bidarra
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 5 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - C C Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 5 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
39
|
Iżycka-Świeszewska E, Gulczyński J, Sejda A, Kitlińska J, Galli S, Rogowski W, Sigorski D. Remarks on Selected Morphological Aspects of Cancer Neuroscience: A Microscopic Photo Review. Biomedicines 2024; 12:2335. [PMID: 39457647 PMCID: PMC11505290 DOI: 10.3390/biomedicines12102335] [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: 08/22/2024] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND This short review and pictorial essay presents a morphological insight into cancer neuroscience, which is a complex and dynamic area of the pathobiology of tumors. METHODS We discuss the different methods and issues connected with structural research on tumor innervation, interactions between neoplastic cells and the nervous system, and dysregulated neural influence on cancer phenotypes. RESULTS Perineural invasion (PNI), the most-visible cancer-nerve relation, is briefly presented, focusing on its pathophysiology and structural diversity as well as its clinical significance. The morphological approach to cancer neurobiology further includes the analysis of neural density/axonogenesis, neural network topographic distribution, and composition of fiber types and size. Next, the diverse range of neurotransmitters and neuropeptides and the neuroendocrine differentiation of cancer cells are reviewed. Another morphological area of cancer neuroscience is spatial or quantitative neural-related marker expression analysis through different detection, description, and visualization methods, also on experimental animal or cellular models. CONCLUSIONS Morphological studies with systematic methodologies provide a necessary insight into the structure and function of the multifaceted tumor neural microenvironment and in context of possible new therapeutic neural-based oncological solutions.
Collapse
Affiliation(s)
- Ewa Iżycka-Świeszewska
- Department of Pathology and Neuropathology, Medical University of Gdansk, 80-210 Gdansk, Poland;
- Department of Pathomorphology, Copernicus Hospital, 80-803 Gdansk, Poland
| | - Jacek Gulczyński
- Department of Pathology and Neuropathology, Medical University of Gdansk, 80-210 Gdansk, Poland;
- Department of Pathomorphology, Copernicus Hospital, 80-803 Gdansk, Poland
| | - Aleksandra Sejda
- Department of Pathomorphology an Forensic Medicine, Collegium Medicum, University of Warmia and Mazury, 10-561 Olsztyn, Poland
| | - Joanna Kitlińska
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA; (J.K.); (S.G.)
| | - Susana Galli
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA; (J.K.); (S.G.)
| | - Wojciech Rogowski
- Institute of Health Sciences, Pomeranian University, 70-204 Slupsk, Poland
| | - Dawid Sigorski
- Department of Oncology, Collegium Medicum, University of Warmia and Mazury, 10-228 Olsztyn, Poland
| |
Collapse
|
40
|
Wilczyński B, Dąbrowska A, Kulbacka J, Baczyńska D. Chemoresistance and the tumor microenvironment: the critical role of cell-cell communication. Cell Commun Signal 2024; 22:486. [PMID: 39390572 PMCID: PMC11468187 DOI: 10.1186/s12964-024-01857-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024] Open
Abstract
Resistance of cancer cells to anticancer drugs remains a major challenge in modern medicine. Understanding the mechanisms behind the development of chemoresistance is key to developing appropriate therapies to counteract it. Nowadays, with advances in technology, we are paying more and more attention to the role of the tumor microenvironment (TME) and intercellular interactions in this process. We also know that important elements of the TME are not only the tumor cells themselves but also other cell types, such as mesenchymal stem cells, cancer-associated fibroblasts, stromal cells, and macrophages. TME elements can communicate with each other indirectly (via cytokines, chemokines, growth factors, and extracellular vesicles [EVs]) and directly (via gap junctions, ligand-receptor pairs, cell adhesion, and tunnel nanotubes). This communication appears to be critical for the development of chemoresistance. EVs seem to be particularly interesting structures in this regard. Within these structures, lipids, proteins, and nucleic acids can be transported, acting as signaling molecules that interact with numerous biochemical pathways, thereby contributing to chemoresistance. Moreover, drug efflux pumps, which are responsible for removing drugs from cancer cells, can also be transported via EVs.
Collapse
Affiliation(s)
- Bartosz Wilczyński
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, Wroclaw, 50-367, Poland
| | - Alicja Dąbrowska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, Wroclaw, 50-367, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, Wroclaw, 50-556, Poland.
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Santariškių g. 5, Vilnius, LT-08406, Lithuania.
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, Wroclaw, 50-556, Poland
| |
Collapse
|
41
|
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
|
42
|
Gebing P, Loizou S, Hänsch S, Schliehe-Diecks J, Spory L, Stachura P, Jepsen VH, Vogt M, Pandyra AA, Wang H, Zhuang Z, Zimmermann J, Schrappe M, Cario G, Alsadeq A, Schewe DM, Borkhardt A, Lenk L, Fischer U, Bhatia S. A brain organoid/ALL coculture model reveals the AP-1 pathway as critically associated with CNS involvement of BCP-ALL. Blood Adv 2024; 8:4997-5011. [PMID: 39008716 PMCID: PMC11465051 DOI: 10.1182/bloodadvances.2023011145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 05/06/2024] [Accepted: 06/26/2024] [Indexed: 07/17/2024] Open
Abstract
ABSTRACT Central nervous system (CNS) involvement remains a clinical hurdle in treating childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL). The disease mechanisms of CNS leukemia are primarily investigated using 2-dimensional cell culture and mouse models. Given the variations in cellular identity and architecture between the human and murine CNS, it becomes imperative to seek complementary models to study CNS leukemia. Here, we present a first-of-its-kind 3-dimensional coculture model combining human brain organoids and BCP-ALL cells. We noticed significantly higher engraftment of BCP-ALL cell lines and patient-derived xenograft (PDX) cells in cerebral organoids than non-ALL cells. To validate translatability between organoid coculture and in vivo murine models, we confirmed that targeting CNS leukemia-relevant pathways such as CD79a/Igα or C-X-C motif chemokine receptor 4-stromal cell-derived factor 1 reduced the invasion of BCP-ALL cells into organoids. RNA sequencing and functional validations of organoid-invading leukemia cells compared with the noninvaded fraction revealed significant upregulation of activator protein 1 (AP-1) transcription factor-complex members in organoid-invading cells. Moreover, we detected a significant enrichment of AP-1 pathway genes in PDX ALL cells recovered from the CNS compared with spleen blasts of mice that had received transplantation with TCF3::PBX1+ PDX cells, substantiating the role of AP-1 signaling in CNS disease. Accordingly, we found significantly higher levels of the AP-1 gene, jun proto-oncogene, in patients initially diagnosed as CNS-positive BCP-ALL compared with CNS-negative cases as well as CNS-relapse vs non-CNS-relapse cases in a cohort of 100 patients with BCP-ALL. Our results suggest CNS organoids as a novel model to investigate CNS involvement and identify the AP-1 pathway as a critical driver of CNS disease in BCP-ALL.
Collapse
Affiliation(s)
- Philip Gebing
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stefanos Loizou
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sebastian Hänsch
- Center for Advanced Imaging, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julian Schliehe-Diecks
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Lea Spory
- Department of Pediatrics I, Pediatric Hematology/Oncology, ALL-BFM Study Group, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Pawel Stachura
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Vera H. Jepsen
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Melina Vogt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Aleksandra A. Pandyra
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Herui Wang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Zhengping Zhuang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Johannes Zimmermann
- Research Group Evolutionary Ecology and Genetics, Zoological Institute, Kiel University, Kiel, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University, Jena, Germany
| | - Martin Schrappe
- Department of Pediatrics I, Pediatric Hematology/Oncology, ALL-BFM Study Group, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Gunnar Cario
- Department of Pediatrics I, Pediatric Hematology/Oncology, ALL-BFM Study Group, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Ameera Alsadeq
- Institute of Immunology, Ulm University Medical Centre, Ulm, Germany
| | - Denis M. Schewe
- Department of Pediatric Hematology and Oncology, University Hospital Dresden, Dresden, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Cancer Consortium, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Lennart Lenk
- Department of Pediatrics I, Pediatric Hematology/Oncology, ALL-BFM Study Group, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Cancer Consortium, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Sanil Bhatia
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Cancer Consortium, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
43
|
Plesselova S, Calar K, Axemaker H, Sahly E, Bhagia A, Faragher JL, Fink DM, de la Puente P. Multicompartmentalized Microvascularized Tumor-on-a-Chip to Study Tumor-Stroma Interactions and Drug Resistance in Ovarian Cancer. Cell Mol Bioeng 2024; 17:345-367. [PMID: 39513004 PMCID: PMC11538101 DOI: 10.1007/s12195-024-00817-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/26/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction The majority of ovarian cancer (OC) patients receiving standard of care chemotherapy develop chemoresistance within 5 years. The tumor microenvironment (TME) is a dynamic and influential player in disease progression and therapeutic response. However, there is a lack of models that allow us to elucidate the compartmentalized nature of TME in a controllable, yet physiologically relevant manner and its critical role in modulating drug resistance. Methods We developed a 3D microvascularized multiniche tumor-on-a-chip formed by five chambers (central cancer chamber, flanked by two lateral stromal chambers and two external circulation chambers) to recapitulate OC-TME compartmentalization and study its influence on drug resistance. Stromal chambers included endothelial cells alone or cocultured with normal fibroblasts or cancer-associated fibroblasts (CAF). Results The tumor-on-a-chip recapitulated spatial TME compartmentalization including vessel-like structure, stromal-mediated extracellular matrix (ECM) remodeling, generation of oxygen gradients, and delayed drug diffusion/penetration from the circulation chamber towards the cancer chamber. The cancer chamber mimicked metastasis-like migration and increased drug resistance to carboplatin/paclitaxel treatment in the presence of CAF when compared to normal fibroblasts. CAF-mediated drug resistance was rescued by ECM targeted therapy. Critically, these results demonstrate that cellular crosstalk recreation and spatial organization through compartmentalization are essential to determining the effect of the compartmentalized OC-TME on drug resistance. Conclusions Our results present a functionally characterized microvascularized multiniche tumor-on-a-chip able to recapitulate TME compartmentalization influencing drug resistance. This technology holds the potential to guide the design of more effective and targeted therapeutic strategies to overcome chemoresistance in OC. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00817-y.
Collapse
Affiliation(s)
- Simona Plesselova
- Present Address: Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD USA
| | - Kristin Calar
- Present Address: Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD USA
| | - Hailey Axemaker
- Present Address: Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD USA
| | - Emma Sahly
- Present Address: Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD USA
- St. Olaf College, Northfield, MN USA
| | - Amrita Bhagia
- MD PhD Program, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD USA
| | - Jessica L. Faragher
- Present Address: Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD USA
- MD PhD Program, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD USA
| | - Darci M. Fink
- Department of Chemistry, Biochemistry & Physics, South Dakota State University, Brookings, SD USA
| | - Pilar de la Puente
- Present Address: Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD USA
- Department of Obstetrics and Gynecology, University of South Dakota Sanford School of Medicine, Sioux Falls, SD USA
- Department of Surgery, University of South Dakota Sanford School of Medicine, Sioux Falls, SD USA
- Flow Cytometry Core, Sanford Research, Sioux Falls, SD USA
| |
Collapse
|
44
|
Gaebler D, Hachey SJ, Hughes CCW. Improving tumor microenvironment assessment in chip systems through next-generation technology integration. Front Bioeng Biotechnol 2024; 12:1462293. [PMID: 39386043 PMCID: PMC11461320 DOI: 10.3389/fbioe.2024.1462293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
The tumor microenvironment (TME) comprises a diverse array of cells, both cancerous and non-cancerous, including stromal cells and immune cells. Complex interactions among these cells play a central role in driving cancer progression, impacting critical aspects such as tumor initiation, growth, invasion, response to therapy, and the development of drug resistance. While targeting the TME has emerged as a promising therapeutic strategy, there is a critical need for innovative approaches that accurately replicate its complex cellular and non-cellular interactions; the goal being to develop targeted, personalized therapies that can effectively elicit anti-cancer responses in patients. Microfluidic systems present notable advantages over conventional in vitro 2D co-culture models and in vivo animal models, as they more accurately mimic crucial features of the TME and enable precise, controlled examination of the dynamic interactions among multiple human cell types at any time point. Combining these models with next-generation technologies, such as bioprinting, single cell sequencing and real-time biosensing, is a crucial next step in the advancement of microfluidic models. This review aims to emphasize the importance of this integrated approach to further our understanding of the TME by showcasing current microfluidic model systems that integrate next-generation technologies to dissect cellular intra-tumoral interactions across different tumor types. Carefully unraveling the complexity of the TME by leveraging next generation technologies will be pivotal for developing targeted therapies that can effectively enhance robust anti-tumoral responses in patients and address the limitations of current treatment modalities.
Collapse
Affiliation(s)
- Daniela Gaebler
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Stephanie J. Hachey
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Christopher C. W. Hughes
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
- Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
45
|
Bhattacharya T, Kumari M, Kaur K, Kaity S, Arumugam S, Ravichandiran V, Roy S. Decellularized extracellular matrix-based bioengineered 3D breast cancer scaffolds for personalized therapy and drug screening. J Mater Chem B 2024; 12:8843-8867. [PMID: 39162395 DOI: 10.1039/d4tb00680a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Breast cancer (BC) is the second deadliest cancer after lung cancer. Similar to all cancers, it is also driven by a 3D microenvironment. The extracellular matrix (ECM) is an essential component of the 3D tumor micro-environment, wherein it functions as a scaffold for cells and provides metabolic support. BC is characterized by alterations in the ECM. Various studies have attempted to mimic BC-specific ECMs using artificial materials, such as Matrigel. Nevertheless, research has proven that naturally derived decellularized extracellular matrices (dECMs) are superior in providing the essential in vivo-like cues needed to mimic a cancer-like environment. Developing in vitro 3-D BC models is not straightforward and requires extensive analysis of the data established by researchers. For the benefit of researchers, in this review, we have tried to highlight all developmental studies that have been conducted by various scientists so far. The analysis of the conclusions drawn from these studies is also discussed. The advantages and drawbacks of the decellularization methods employed for generating BC scaffolds will be covered, and the review will shed light on how dECM scaffolds help develop a BC environment. The later stages of the article will also focus on immunogenicity issues arising from decellularization and the origin of the tissue. Finally, this review will also discuss the biofabrication of matrices, which is the core part of the bioengineering process.
Collapse
Affiliation(s)
- Teeshyo Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| | - Mamta Kumari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Somasundaram Arumugam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| | - Velayutham Ravichandiran
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| |
Collapse
|
46
|
Shiraishi A, Oh-Hara T, Takahashi Y, Uchibori K, Nishio M, Katayama R. 3D layered co-culture model enhances Trastuzumab Deruxtecan sensitivity and reveals the combined effect with G007-LK in HER2-positive non-small cell lung cancer. Biochem Biophys Res Commun 2024; 725:150255. [PMID: 38897043 DOI: 10.1016/j.bbrc.2024.150255] [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: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
Human epidermal growth factor receptor 2 (HER2) aberrations are observed in various cancers. In non-small cell lung cancer, genetic alterations activating HER2, mostly exon 20 insertion mutations, occur in approximately 2-4% of cases. Trastuzumab deruxtecan (T-DXd), a HER2-targeted antibody-drug conjugate has been approved as the first HER2-targeted drug for HER2-mutant lung cancer. However, some cases are not responsive to T-DXd and the primary resistant mechanism remains unclear. In this study, we assessed sensitivity to T-DXd in JFCR-007, a patient-derived HER2-mutant lung cancer cell line. Although JFCR-007 was sensitive to HER2 tyrosine kinase inhibitors, it showed resistance to T-DXd in attachment or spheroid conditions. Accordingly, we established a three-dimensional (3D) layered co-culture model of JFCR-007, where it exhibited a lumen-like structure and became sensitive to T-DXd. In addition, an in-house inhibitor library screening revealed that G007-LK, a tankyrase inhibitor, was effective when combined with T-DXd. G007-LK increased the cytotoxicity of topoisomerase-I inhibitor, DXd, a payload of T-DXd and SN-38. This combined effect was also observed in H2170, an HER2-amplified lung cancer cell line. These results suggest that the proposed 3D co-culture system may help in evaluating the efficacy of T-DXd and may recapitulate the tumor microenvironment.
Collapse
MESH Headings
- Humans
- Trastuzumab/pharmacology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Cell Line, Tumor
- Immunoconjugates/pharmacology
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Coculture Techniques
- Drug Resistance, Neoplasm/drug effects
- Crown Ethers/pharmacology
- Antineoplastic Agents, Immunological/pharmacology
- Camptothecin/analogs & derivatives
Collapse
Affiliation(s)
- Akari Shiraishi
- Div of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa-shi, Chiba, 277-8561, Japan
| | - Tomoko Oh-Hara
- Div of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Yuki Takahashi
- Technical Research Institute, TOPPAN Holdings Inc., Japan; Division of Clinical Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Ken Uchibori
- Div of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan; Department of Thoracic Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makoto Nishio
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryohei Katayama
- Div of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa-shi, Chiba, 277-8561, Japan.
| |
Collapse
|
47
|
Lampejo AO, Lightsey SE, Gomes MC, Nguyen CM, Siemann DW, Sharma B, Murfee WL. A Novel Ex Vivo Tumor Spheroid-Tissue Model for Investigating Microvascular Remodeling and Lymphatic Blood Vessel Plasticity. Ann Biomed Eng 2024; 52:2457-2472. [PMID: 38796670 DOI: 10.1007/s10439-024-03535-8] [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: 12/12/2023] [Accepted: 05/02/2024] [Indexed: 05/28/2024]
Abstract
Biomimetic tumor microenvironment models bridge the gap between in vitro and in vivo systems and serve as a useful way to address the modeling challenge of how to recreate the cell and system complexity associated with real tissues. Our laboratory has developed an ex vivo rat mesentery culture model, which allows for simultaneous investigation of blood and lymphatic microvascular network remodeling in an intact tissue environment. Given that angiogenesis and lymphangiogenesis are key contributors to the progression of cancer, the objective of this study was to combine tissue and tumor spheroid culture methods to establish a novel ex vivo tumor spheroid-tissue model by verifying its use for evaluating the effects of cancer cell behavior on the local microvascular environment. H1299 or A549 tumor spheroids were formed via hanging drop culture and seeded onto rat mesenteric tissues harvested from adult male Wistar rats. Tissues with transplanted spheroids were cultured in serum-free media for 3 to 5 days. PECAM, NG2, CD11b, and αSMA labeling identified endothelial cells, pericytes, immune cells, and smooth muscle cells, respectively. Time-lapse imaging confirmed cancer cell type specific migration. In addition to increasing PECAM positive capillary sprouting and LYVE-1 positive endothelial cell extensions indicative of lymphangiogenesis, tumor spheroid presence induced the formation of lymphatic/blood vessel connections and the formation of hybrid, mosaic vessels that were characterized by discontinuous LYVE-1 labeling. The results support the application of a novel tumor spheroid microenvironment model for investigating cancer cell-microvascular interactions.
Collapse
Affiliation(s)
- Arinola O Lampejo
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Suzanne E Lightsey
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Maria C Gomes
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Christian M Nguyen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar W Siemann
- University of Florida Health Cancer Center, Gainesville, FL, USA
- Department of Radiation Oncology, University of Florida, University of Florida Health, Gainesville, USA
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Walter L Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- University of Florida Health Cancer Center, Gainesville, FL, USA.
| |
Collapse
|
48
|
Aydin H, Ozcelikkale A, Acar A. Exploiting Matrix Stiffness to Overcome Drug Resistance. ACS Biomater Sci Eng 2024; 10:4682-4700. [PMID: 38967485 PMCID: PMC11322920 DOI: 10.1021/acsbiomaterials.4c00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
Drug resistance is arguably one of the biggest challenges facing cancer research today. Understanding the underlying mechanisms of drug resistance in tumor progression and metastasis are essential in developing better treatment modalities. Given the matrix stiffness affecting the mechanotransduction capabilities of cancer cells, characterization of the related signal transduction pathways can provide a better understanding for developing novel therapeutic strategies. In this review, we aimed to summarize the recent advancements in tumor matrix biology in parallel to therapeutic approaches targeting matrix stiffness and its consequences in cellular processes in tumor progression and metastasis. The cellular processes governed by signal transduction pathways and their aberrant activation may result in activating the epithelial-to-mesenchymal transition, cancer stemness, and autophagy, which can be attributed to drug resistance. Developing therapeutic strategies to target these cellular processes in cancer biology will offer novel therapeutic approaches to tailor better personalized treatment modalities for clinical studies.
Collapse
Affiliation(s)
- Hakan
Berk Aydin
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| | - Altug Ozcelikkale
- Department
of Mechanical Engineering, Middle East Technical
University, 06800, Ankara, Turkey
- Graduate
Program of Biomedical Engineering, Middle
East Technical University, 06800, Ankara, Turkey
| | - Ahmet Acar
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| |
Collapse
|
49
|
Prakash J, Shaked Y. The Interplay between Extracellular Matrix Remodeling and Cancer Therapeutics. Cancer Discov 2024; 14:1375-1388. [PMID: 39091205 PMCID: PMC11294818 DOI: 10.1158/2159-8290.cd-24-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/12/2024] [Accepted: 05/22/2024] [Indexed: 08/04/2024]
Abstract
The extracellular matrix (ECM) is an abundant noncellular component of most solid tumors known to support tumor progression and metastasis. The interplay between the ECM and cancer therapeutics opens up new avenues in understanding cancer biology. While the ECM is known to protect the tumor from anticancer agents by serving as a biomechanical barrier, emerging studies show that various cancer therapies induce ECM remodeling, resulting in therapy resistance and tumor progression. This review discusses critical issues in this field including how the ECM influences treatment outcome, how cancer therapies affect ECM remodeling, and the challenges associated with targeting the ECM. Significance: The intricate relationship between the extracellular matrix (ECM) and cancer therapeutics reveals novel insights into tumor biology and its effective treatment. While the ECM may protect tumors from anti-cancer agents, recent research highlights the paradoxical role of therapy-induced ECM remodeling in promoting treatment resistance and tumor progression. This review explores the key aspects of the interplay between ECM and cancer therapeutics.
Collapse
Affiliation(s)
- Jai Prakash
- Engineered Therapeutics Group, Department of Advanced Organ Bioengineering and Therapeutics, Technical Medical Centre, University of Twente, Enschede, the Netherlands.
| | - Yuval Shaked
- Rappaport Faculty of Medicine, Rappaport-Technion Integrated Cancer Center, Technion – Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
50
|
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
|