1
|
Tang R, Luo S, Liu H, Sun Y, Liu M, Li L, Ren H, Angele MK, Börner N, Yu K, Guo Z, Yin G, Luo H. Circulating Tumor Microenvironment in Metastasis. Cancer Res 2025; 85:1354-1367. [PMID: 39992721 PMCID: PMC11997552 DOI: 10.1158/0008-5472.can-24-1241] [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: 04/15/2024] [Revised: 11/12/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Activation of invasion and metastasis is a central hallmark of cancer, contributing to the primary cause of death for patients with cancer. In the multistep metastatic process, cancer cells must infiltrate the circulation, survive, arrest at capillary beds, extravasate, and form metastatic clones in distant organs. However, only a small proportion of circulating tumor cells (CTC) successfully form metastases, with transit of CTCs in the circulation being the rate-limiting step. The fate of CTCs is influenced by the circulating tumor microenvironment (cTME), which encompasses factors affecting their biological behaviors in the circulation. This liquid and flowing microenvironment differs significantly from the primary TME or the premetastatic niche. This review summarizes the latest advancements in identifying the biophysical cues, key components, and biological roles of the cTME, highlighting the network among biophysical attributes, blood cells, and nonblood factors in cancer metastasis. In addition to the potential of the cTME as a therapeutic target for inhibiting metastasis, the cTME could also represent as a biomarker for predicting patient outcomes and developing strategies for treating cancer.
Collapse
Affiliation(s)
- Rui Tang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Shujuan Luo
- Department of Obstetrics, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Liu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yan Sun
- Department of Cell Biology and Medical Genetics, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Lu Li
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haoyu Ren
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Martin K. Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich Munich, Germany
| | - Nikolaus Börner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich Munich, Germany
| | - Keda Yu
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Zufeng Guo
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Guobing Yin
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haojun Luo
- Department of Thyroid and Breast Surgery, Renji Hospital, School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
2
|
Devenport JM, Tran T, Harris BR, Fingerman D, DeWeerd RA, Elkhidir LH, LaVigne D, Fuh K, Sun L, Bednarski JJ, Drapkin R, Mullen MM, Green AM. APOBEC3A drives ovarian cancer metastasis by altering epithelial-mesenchymal transition. JCI Insight 2025; 10:e186409. [PMID: 40059825 PMCID: PMC11949045 DOI: 10.1172/jci.insight.186409] [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: 08/28/2024] [Accepted: 01/22/2025] [Indexed: 03/19/2025] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is the most prevalent and aggressive histological subtype of ovarian cancer and often presents with metastatic disease. The drivers of metastasis in HGSOC remain enigmatic. APOBEC3A (A3A), an enzyme that generates mutations across various cancers, has been proposed as a mediator of tumor heterogeneity and disease progression. However, the role of A3A in HGSOC has not been explored. We observed an association between high levels of APOBEC3-mediated mutagenesis and poor overall survival in primary HGSOC. We experimentally addressed this correlation by modeling A3A expression in HGSOC, and this resulted in increased metastatic behavior of HGSOC cells in culture and distant metastatic spread in vivo, which was dependent on catalytic activity of A3A. A3A activity in both primary and cultured HGSOC cells yielded consistent alterations in expression of epithelial-mesenchymal transition (EMT) genes resulting in hybrid EMT and mesenchymal signatures, providing a mechanism for their increased metastatic potential. Inhibition of key EMT factors TWIST1 and IL-6 resulted in mitigation of A3A-dependent metastatic phenotypes. Our findings define the prevalence of A3A mutagenesis in HGSOC and implicate A3A as a driver of HGSOC metastasis via EMT, underscoring its clinical relevance as a potential prognostic biomarker. Our study lays the groundwork for the development of targeted therapies aimed at mitigating the deleterious effect of A3A-driven EMT in HGSOC.
Collapse
Affiliation(s)
| | | | | | - Dylan Fingerman
- Department of Pediatrics
- Cancer Biology Graduate Program, and
| | | | | | - Danielle LaVigne
- Department of Pediatrics
- Molecular Genetics and Genomics Graduate Program, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Katherine Fuh
- Department of Obstetrics, Gynecology, and Reproductive Sciences, UCSF, San Francisco, California, USA
| | - Lulu Sun
- Division of Anatomic and Molecular Pathology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, and
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Mary M. Mullen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Siteman Cancer Center, and
| | - Abby M. Green
- Department of Pediatrics
- Center for Genome Integrity, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
3
|
Li CX, Zhao ZX, Su DB, Yin DC, Ye YJ. In vitro regulation of collective cell migration: Understanding the role of physical and chemical microenvironments. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:23-40. [PMID: 39612952 DOI: 10.1016/j.pbiomolbio.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/01/2024]
Abstract
Collective cell migration is the primary mode of cellular movement during embryonic morphogenesis, tissue repair and regeneration, and cancer invasion. Distinct from single-cell migration, collective cell migration involves complex intercellular signaling cascades and force transmission. Consequently, cell collectives exhibit intricate and diverse migration patterns under the influence of the microenvironment in vivo. Investigating the patterns and mechanisms of collective cell migration within complex environmental factors in vitro is essential for elucidating collective cell migration in vivo. This review elucidates the influence of physical and chemical factors in vitro microenvironment on the migration patterns and efficiency of cell collectives, thereby enhancing our comprehension of the phenomenon. Furthermore, we concisely present the effects of characteristic properties of common biomaterials on collective cell migration during tissue repair and regeneration, as well as the features and applications of tumor models of different dimensions (2D substrate or 3D substrate) in vitro. Finally, we highlight the challenges facing the research of collective cell migration behaviors in vitro microenvironment and propose that modulating collective cell migration may represent a potential strategy to promote tissue repair and regeneration and to control tumor invasion and metastasis.
Collapse
Affiliation(s)
- Chang-Xing Li
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Zi-Xu Zhao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Dan-Bo Su
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Da-Chuan Yin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Ya-Jing Ye
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
4
|
Thibault B, Thole A, D'Angelo R, Basset C, Guillermet-Guibert J. PI3Kα-specific inhibitor BYL-719 synergizes with cisplatin in vitro in PIK3CA-mutated ovarian cancer cells. Sci Rep 2025; 15:6265. [PMID: 39979449 PMCID: PMC11842864 DOI: 10.1038/s41598-025-90714-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/14/2025] [Indexed: 02/22/2025] Open
Abstract
Peritoneal carcinomatosis in ovarian cancer is often associated with ascites where cancer cells grow as aggregates. Given the emerging evidence that multicellular growth enhances resistance to conventional therapies, and that patients frequently develop resistance to platinum salts, we investigated the efficiency of PI3K/Akt signalling pathway targeting in multicellular growth and its importance as a potential therapeutic target in cells resistant to platinum salts. Due to its importance in many cancers and to the frequent mutations of its encoding gene PIK3CA, we focused on targeting PI3Kα using BYL-719 (Alpelisib), an isoform-specific inhibitor already used in clinics. We used a panel of 3 ovarian cancer cell lines, SKOV-3, EFO-21 and OVCAR-3, which come from different histological origins and bear different mutations. PI3K targeting drugs inhibit the activity of the PI3K/Akt pathway in all tested ovarian cancer cell lines with a drastic reduction of the phosphorylation of Akt on the serine 473, regardless the histology or the mutational profile. We showed that when cultured in 3D aggregates, ovarian cancer cells are more resistant to the PI3Kα-specific inhibitor BYL-719 and cisplatin compared to 2D monolayers. BYL-719 synergizes with cisplatin in 3D cultures only in PIK3CA-mutated SKOV-3 cells. This drug combination leads to a major cytotoxicity in 3D aggregates of this cell line. Finally, BYL-719 in combination with cisplatin remains active in 3D aggregates of SKOV-3 cells co-cultured with mesenchymal stem cells. We have identified a signalling pathway of interest for the treatment of advanced ovarian cancer in vitro, which could limit the progression of this disease. These data pave the road to investigate whether PI3Kα-specific inhibitor BYL-719 should be proposed in combination with cisplatin, in priority in patients bearing a PIK3CA mutation.
Collapse
Affiliation(s)
- Benoît Thibault
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, Equipe SigDYN, 2 av Hubert Curien, Oncopole de Toulouse, Toulouse, France.
- Labex TOUCAN, ANR, Toulouse, France.
| | - Adrien Thole
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, Equipe SigDYN, 2 av Hubert Curien, Oncopole de Toulouse, Toulouse, France
- Labex TOUCAN, ANR, Toulouse, France
| | - Romina D'Angelo
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, Equipe SigDYN, 2 av Hubert Curien, Oncopole de Toulouse, Toulouse, France
- Labex TOUCAN, ANR, Toulouse, France
| | - Céline Basset
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, Equipe SigDYN, 2 av Hubert Curien, Oncopole de Toulouse, Toulouse, France
- Labex TOUCAN, ANR, Toulouse, France
- Service d'Anatomo-Pathologie, Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), avenue Irène Joliot-Curie, Toulouse, France
| | - Julie Guillermet-Guibert
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, Equipe SigDYN, 2 av Hubert Curien, Oncopole de Toulouse, Toulouse, France.
- Labex TOUCAN, ANR, Toulouse, France.
| |
Collapse
|
5
|
Mehta G, Horst E, Cotter L, Bonini M, Novak C, Treacher N, Zhang Y, Jackson Z, Narayanan IV, Wuchu F, Nenwani M, Fischer Z, Sunshine A, Lin Z, Tran L, Nagrath D, Ljungman M, Maturen K, DiFeo A, Nordsletten D. Ascitic Shear Stress Activates GPCRs and Downregulates Mucin 15 to Promote Ovarian Cancer Malignancy. RESEARCH SQUARE 2024:rs.3.rs-5160301. [PMID: 39483899 PMCID: PMC11527234 DOI: 10.21203/rs.3.rs-5160301/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The accumulation of ascites in patients with ovarian cancer increases their risk of transcoelomic metastasis. Although common routes of peritoneal dissemination are known to follow distinct paths of circulating ascites, the mechanisms that initiate these currents and subsequent fluid shear stresses are not well understood. Here, we developed a patient-based, boundary-driven computational fluid dynamics model to predict an upper range of fluid shear stress generated by the accumulation of ascites. We show that ovarian cancer cells exposed to ascitic shear stresses display heightened G protein-coupled receptor mechanosignaling and the induction of an epithelial to mesenchymal-like transition through p38α mitogen-activated protein kinase and mucin 15 modulation. These findings along with a shear-induced immunomodulatory secretome position elevated shear stress as a protumoural signal. Together, this study suggests inhibition of the Gαq protein and restriction of ascites accumulation as maintenance strategies for overcoming mechanotransduction-mediated metastasis within the peritoneal cavity.
Collapse
|
6
|
Ruhi MK, Rickard BP, Overchuk M, Sinawang PD, Stanley E, Mansi M, Sierra RG, Hayes B, Tan X, Akin D, Chen B, Demirci U, Rizvi I. PpIX-enabled fluorescence-based detection and photodynamic priming of platinum-resistant ovarian cancer cells under fluid shear stress. Photochem Photobiol 2024; 100:1603-1621. [PMID: 39189505 DOI: 10.1111/php.14014] [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: 03/28/2024] [Revised: 06/27/2024] [Accepted: 06/30/2024] [Indexed: 08/28/2024]
Abstract
Over 75% percent of ovarian cancer patients are diagnosed with advanced-stage disease characterized by unresectable intraperitoneal dissemination and the presence of ascites, or excessive fluid build-up within the abdomen. Conventional treatments include cytoreductive surgery followed by multi-line platinum and taxane chemotherapy regimens. Despite an initial response to treatment, over 75% of patients with advanced-stage ovarian cancer will relapse and succumb to platinum-resistant disease. Recent evidence suggests that fluid shear stress (FSS), which results from the movement of fluid such as ascites, induces epithelial-to-mesenchymal transition and confers resistance to carboplatin in ovarian cancer cells. This study demonstrates, for the first time, that FSS-induced platinum resistance correlates with increased cellular protoporphyrin IX (PpIX), the penultimate downstream product of heme biosynthesis, the production of which can be enhanced using the clinically approved pro-drug aminolevulinic acid (ALA). These data suggest that, with further investigation, PpIX could serve as a fluorescence-based biomarker of FSS-induced platinum resistance. Additionally, this study investigates the efficacy of PpIX-enabled photodynamic therapy (PDT) and the secretion of extracellular vesicles under static and FSS conditions in Caov-3 and NIH:OVCAR-3 cells, two representative cell lines for high-grade serous ovarian carcinoma (HGSOC), the most lethal form of the disease. FSS induces resistance to ALA-PpIX-mediated PDT, along with a significant increase in the number of EVs. Finally, the ability of PpIX-mediated photodynamic priming (PDP) to enhance carboplatin efficacy under FSS conditions is quantified. These preliminary findings in monolayer cultures necessitate additional studies to determine the feasibility of PpIX as a fluorescence-based indicator, and mediator of PDP, to target chemoresistance in the context of FSS.
Collapse
Affiliation(s)
- Mustafa Kemal Ruhi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Brittany P Rickard
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Marta Overchuk
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
| | - Prima Dewi Sinawang
- Department of Chemical Engineering, School of Engineering, Stanford University, Stanford, California, USA
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratories, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California, USA
- Department of Radiology, School of Medicine, Canary Center at Stanford, Stanford University, Palo Alto, California, USA
| | - Elizabeth Stanley
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
| | - Matthew Mansi
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph's University, Philadelphia, Pennsylvania, USA
| | - Raymond G Sierra
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, Menlo Park, California, USA
| | - Brandon Hayes
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, Menlo Park, California, USA
| | - Xianming Tan
- Department of Biostatistics, University of North Carolina School of Public Health, Chapel Hill, North Carolina, USA
| | - Demir Akin
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratories, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California, USA
- Department of Radiology, School of Medicine, Canary Center at Stanford, Stanford University, Palo Alto, California, USA
| | - Bin Chen
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph's University, Philadelphia, Pennsylvania, USA
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratories, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California, USA
- Department of Radiology, School of Medicine, Canary Center at Stanford, Stanford University, Palo Alto, California, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
7
|
Overchuk M, Rickard BP, Tulino J, Tan X, Ligler FS, Huang HC, Rizvi I. Overcoming the effects of fluid shear stress in ovarian cancer cell lines: Doxorubicin alone or photodynamic priming to target platinum resistance. Photochem Photobiol 2024; 100:1676-1693. [PMID: 38849970 PMCID: PMC11568959 DOI: 10.1111/php.13967] [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: 01/03/2024] [Revised: 04/15/2024] [Accepted: 05/05/2024] [Indexed: 06/09/2024]
Abstract
Resistance to platinum-based chemotherapies remains a significant challenge in advanced-stage high-grade serous ovarian carcinoma, and patients with malignant ascites face the poorest outcomes. It is, therefore, important to understand the effects of ascites, including the associated fluid shear stress (FSS), on phenotypic changes and therapy response, specifically FSS-induced chemotherapy resistance and the underlying mechanisms in ovarian cancer. This study investigated the effects of FSS on response to cisplatin, a platinum-based chemotherapy, and doxorubicin, an anthracycline, both of which are commonly used to manage advanced-stage ovarian cancer. Consistent with prior research, OVCAR-3 and Caov-3 cells cultivated under FSS demonstrated significant resistance to cisplatin. Examination of the role of mitochondria revealed an increase in mitochondrial DNA copy number and intracellular ATP content in cultures grown under FSS, suggesting that changes in mitochondria number and metabolic activity may contribute to platinum resistance. Interestingly, no resistance to doxorubicin was observed under FSS, the first such observation of a lack of resistance under these conditions. Finally, this study demonstrated the potential of photodynamic priming using benzoporphyrin derivative, a clinically approved photosensitizer that localizes in part to mitochondria and endoplasmic reticula, to enhance the efficacy of cisplatin, but not doxorubicin, thereby overcoming FSS-induced platinum resistance.
Collapse
Affiliation(s)
- Marta Overchuk
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; North Carolina State University, Raleigh, NC 27606, USA
| | - Brittany P. Rickard
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Justin Tulino
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; North Carolina State University, Raleigh, NC 27606, USA
| | - Xianming Tan
- Department of Biostatistics, University of North Carolina School of Public Health, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Frances S. Ligler
- Department of Biomedical Engineering, Texas A&M University, Collage Station, TX, 77843 USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; North Carolina State University, Raleigh, NC 27606, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
8
|
Scott AL, Jazwinska DE, Kulawiec DG, Zervantonakis IK. Paracrine Ovarian Cancer Cell-Derived CSF1 Signaling Regulates Macrophage Migration Dynamics in a 3D Microfluidic Model that Recapitulates In Vivo Infiltration Patterns in Patient-Derived Xenografts. Adv Healthc Mater 2024; 13:e2401719. [PMID: 38807270 PMCID: PMC11560735 DOI: 10.1002/adhm.202401719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Indexed: 05/30/2024]
Abstract
A high density of macrophages in the ovarian cancer microenvironment is associated with disease progression and poor outcomes. Understanding cancer-macrophage interaction mechanisms that establish this pro-tumorigenic microenvironment is critical for developing macrophage-targeted therapies. Here, 3D microfluidic assays and patient-derived xenografts are utilized to define the role of cancer-derived colony stimulating factor 1 (CSF1) on macrophage infiltration dynamics toward ovarian cancer cells. It is demonstrated that multiple ovarian cancer models promote the infiltration of macrophages into a 3D extracellular matrix in vitro in a cell density-dependent manner. Macrophages exhibit directional migration and increased migration speed under both direct interactions with cancer cells embedded within the matrix and paracrine crosstalk with cancer cells seeded in an independent microchannel. It is also found that platinum-based chemotherapy increases macrophage recruitment and the levels of cancer cell-derived CSF1. Targeting CSF1 signaling under baseline or chemotherapy-treatment conditions reduces the number of infiltrated macrophages. It is further shown that results obtained with the 3D microfluidic model reflect the recruitment profiles of macrophages in patient-derived xenografts in vivo. These findings highlight the role of CSF1 signaling in establishing macrophage-rich ovarian cancer microenvironments, as well as the utility of microfluidic models in recapitulating 3D tumor ecosystems and dissecting cancer-macrophage signaling.
Collapse
Affiliation(s)
- Alexis L Scott
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Dorota E Jazwinska
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Diana G Kulawiec
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Ioannis K Zervantonakis
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| |
Collapse
|
9
|
Pajic-Lijakovic I, Milivojevic M, McClintock PVE. Physical aspects of epithelial cell-cell interactions: hidden system complexities. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2024; 53:355-372. [PMID: 39256261 PMCID: PMC11560995 DOI: 10.1007/s00249-024-01721-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/23/2024] [Accepted: 08/31/2024] [Indexed: 09/12/2024]
Abstract
The maintenance of homeostasis and the retention of ordered epithelial cell self-organization are essential for morphogenesis, wound healing, and the spread of cancer across the epithelium. However, cell-cell interactions in an overcrowded environment introduce a diversity of complications. Such interactions arise from an interplay between the cell compressive and shear stress components that accompany increased cell packing density. They can lead to various kinds of cell rearrangement such as: the epithelial-to-mesenchymal cell state transition; live cell extrusion; and cell jamming. All of these scenarios of cell rearrangement under mechanical stress relate to changes in the strengths of the cell-cell and cell-matrix adhesion contacts. The objective of this review study is twofold: first, to provide a comprehensive summary of the biological and physical factors influencing the effects of cell mechanical stress on cell-cell interactions, and the consequences of these interactions for the status of cell-cell and cell-matrix adhesion contacts; and secondly, to offer a bio-physical/mathematical analysis of the aforementioned biological aspects. By presenting these two approaches in conjunction, we seek to highlight the intricate nature of biological systems, which manifests in the form of complex bio-physical/mathematical equations. Furthermore, the juxtaposition of these apparently disparate approaches underscores the importance of conducting experiments to determine the multitude of parameters that contribute to the development of these intricate bio-physical/mathematical models.
Collapse
Affiliation(s)
- Ivana Pajic-Lijakovic
- Faculty of Technology and Metallurgy, Department of Chemical Engineering, University of Belgrade, Belgrade, Serbia.
| | - Milan Milivojevic
- Faculty of Technology and Metallurgy, Department of Chemical Engineering, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
10
|
Devenport JM, Tran T, Harris BR, Fingerman DF, DeWeerd RA, Elkhidir L, LaVigne D, Fuh K, Sun L, Bednarski JJ, Drapkin R, Mullen M, Green AM. APOBEC3A drives metastasis of high-grade serous ovarian cancer by altering epithelial-to-mesenchymal transition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620297. [PMID: 39553968 PMCID: PMC11565781 DOI: 10.1101/2024.10.25.620297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most prevalent and aggressive histological subtype of ovarian cancer, and often presents with metastatic disease. The drivers of metastasis in HGSOC remain enigmatic. APOBEC3A (A3A), an enzyme that generates mutations across various cancers, has been proposed as a mediator of tumor heterogeneity and disease progression. However, the role of A3A in HGSOC has not been explored. Through analysis of genome sequencing from primary HGSOC, we observed an association between high levels of APOBEC3 mutagenesis and poor overall survival. We experimentally addressed this correlation by modeling A3A activity in HGSOC cell lines and mouse models which resulted in increased metastatic behavior of HGSOC cells in culture and distant metastatic spread in vivo . A3A activity in both primary and cultured HGSOC cells yielded consistent alterations in expression of epithelial-mesenchymal-transition (EMT) genes resulting in hybrid EMT and mesenchymal signatures, and providing a mechanism for their increased metastatic potential. Our findings define the prevalence of A3A mutagenesis in HGSOC and implicate A3A as a driver of HGSOC metastasis via EMT, underscoring its clinical relevance as a potential prognostic biomarker. Our study lays the groundwork for the development of targeted therapies aimed at mitigating the deleterious impact of A3A-driven EMT in HGSOC.
Collapse
|
11
|
Crouigneau R, Li YF, Auxillos J, Goncalves-Alves E, Marie R, Sandelin A, Pedersen SF. Mimicking and analyzing the tumor microenvironment. CELL REPORTS METHODS 2024; 4:100866. [PMID: 39353424 PMCID: PMC11573787 DOI: 10.1016/j.crmeth.2024.100866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 07/22/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024]
Abstract
The tumor microenvironment (TME) is increasingly appreciated to play a decisive role in cancer development and response to therapy in all solid tumors. Hypoxia, acidosis, high interstitial pressure, nutrient-poor conditions, and high cellular heterogeneity of the TME arise from interactions between cancer cells and their environment. These properties, in turn, play key roles in the aggressiveness and therapy resistance of the disease, through complex reciprocal interactions between the cancer cell genotype and phenotype, and the physicochemical and cellular environment. Understanding this complexity requires the combination of sophisticated cancer models and high-resolution analysis tools. Models must allow both control and analysis of cellular and acellular TME properties, and analyses must be able to capture the complexity at high depth and spatial resolution. Here, we review the advantages and limitations of key models and methods in order to guide further TME research and outline future challenges.
Collapse
Affiliation(s)
- Roxane Crouigneau
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Yan-Fang Li
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Jamie Auxillos
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Eliana Goncalves-Alves
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Rodolphe Marie
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Albin Sandelin
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| | - Stine Falsig Pedersen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
12
|
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
|
13
|
Li J, Zhou M, Xie J, Chen J, Yang M, Ye C, Cheng S, Liu M, Li R, Tan R. Organoid modeling meets cancers of female reproductive tract. Cell Death Discov 2024; 10:410. [PMID: 39333482 PMCID: PMC11437045 DOI: 10.1038/s41420-024-02186-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
Diseases of the female reproductive system, especially malignant tumors, pose a serious threat to women's health worldwide. One of the key factors limiting research progress in this area is the lack of representative models. Organoid technology, especially tumor organoids, has been increasingly applied in the study of female reproductive system tumors due to their high heterogeneity, close resemblance to the physiological state, easy acquisition and cultivation advantages. They play a significant role in understanding the origin and causes of tumors, drug screening, and personalized treatment and more. This article reviews the organoid models for the female reproductive system, focusing on the cancer research advancements. It discusses the methods for constructing tumor organoids of the female reproductive tract and summarizes the limitations of current research. The aim is to offer a reference for future development and application of these organoid models, contributing to the advancement of anti-tumor drugs and treatment strategies for female reproductive tract cancer patients.
Collapse
Affiliation(s)
- Jiao Li
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Mengting Zhou
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Xie
- Information Technology Center, West China Hospital of Sichuan University, Sichuan University, Chengdu, China
| | - Jiani Chen
- Chongqing Medical University, Chongqing, China
| | - Mengni Yang
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Changjun Ye
- Rehabilitation Department, Changgeng Yining Hospital, Wenzhou, China
| | - Shihu Cheng
- Geriatric Department, Changgeng Yining Hospital, Wenzhou, China
| | - Miao Liu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| |
Collapse
|
14
|
Pyne E, Reardon M, Christensen M, Rodriguez Mateos P, Taylor S, Iles A, Choudhury A, Pamme N, Pires IM. Investigating the impact of the interstitial fluid flow and hypoxia interface on cancer transcriptomes using a spheroid-on-chip perfusion system. LAB ON A CHIP 2024; 24:4609-4622. [PMID: 39258507 PMCID: PMC11388701 DOI: 10.1039/d4lc00512k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024]
Abstract
Solid tumours are complex and heterogeneous systems, which exist in a dynamic biophysical microenvironment. Conventional cancer research methods have long relied on two-dimensional (2D) static cultures which neglect the dynamic, three-dimensional (3D) nature of the biophysical tumour microenvironment (TME), especially the role and impact of interstitial fluid flow (IFF). To address this, we undertook a transcriptome-wide analysis of the impact of IFF-like perfusion flow using a spheroid-on-chip microfluidic platform, which allows 3D cancer spheroids to be integrated into extracellular matrices (ECM)-like hydrogels and exposed to continuous perfusion, to mimic IFF in the TME. Importantly, we have performed these studies both in experimental (normoxia) and pathophysiological (hypoxia) oxygen conditions. Our data indicated that gene expression was altered by flow when compared to static conditions, and for the first time showed that these gene expression patterns differed in different oxygen tensions, reflecting a differential role of spheroid perfusion in IFF-like flow in tumour-relevant hypoxic conditions in the biophysical TME. We were also able to identify factors primarily linked with IFF-like conditions which are linked with prognostic value in cancer patients and therefore could correspond to a potential novel biomarker of IFF in cancer. This study therefore highlights the need to consider relevant oxygen conditions when studying the impact of flow in cancer biology, as well as demonstrating the potential of microfluidic models of flow to identify IFF-relevant tumour biomarkers.
Collapse
Affiliation(s)
- Emily Pyne
- Centre for Biomedicine, HYMS, University of Hull, Hull, UK
| | - Mark Reardon
- Translational Radiobiology, Division of Cancer Sciences, University of Manchester, Manchester, UK
| | | | - Pablo Rodriguez Mateos
- Department of Materials and Environmental Chemistry, Stockholm University, Stockholm, Sweden
| | - Scott Taylor
- Tumour Hypoxia Biology, Division of Cancer Sciences, University of Manchester, Manchester, UK.
| | - Alexander Iles
- Department of Materials and Environmental Chemistry, Stockholm University, Stockholm, Sweden
| | - Ananya Choudhury
- Translational Radiobiology, Division of Cancer Sciences, University of Manchester, Manchester, UK
- Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Nicole Pamme
- Department of Materials and Environmental Chemistry, Stockholm University, Stockholm, Sweden
| | - Isabel M Pires
- Centre for Biomedicine, HYMS, University of Hull, Hull, UK
- Tumour Hypoxia Biology, Division of Cancer Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
15
|
Chavoshinezhad N, Niknafs B. Innovations in 3D ovarian and follicle engineering for fertility preservation and restoration. Mol Biol Rep 2024; 51:1004. [PMID: 39305382 DOI: 10.1007/s11033-024-09783-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/05/2024] [Indexed: 02/06/2025]
Abstract
In-vitro maturation (IVM) is the process of cultivating early-stage follicles from the primordial to the antral stage and facilitating the maturation of oocytes outside the body within a supportive environment. This intricate procedure requires the careful coordination of various factors to replicate the natural ovarian conditions. Advanced techniques for IVM are designed to mimic the natural ovarian environment and enhance the development of follicles. Three-dimensional (3D) culture systems provide a more biologically relevant setting for follicle growth compared to traditional two-dimensional (2D) cultures. Traditional culture systems, often fail to support the complex process of follicle development effectively. However, modern engineered reproductive tissues and culture systems are making it possible to create increasingly physiological in-vitro models of folliculogenesis. These innovative methods are enabling researchers and clinicians to better replicate the dynamic and supportive environment of the ovary, thereby improving the outcomes of IVM offering new hope for fertility preservation and treatment. This paper focuses on the routine 3D culture, and innovative 3D culture of ovary and follicles, including a tissue engineering scaffolds, microfluidic (dynamic) culture system, organ-on-chip models, EVATAR system, from a clinical perspective to determine the most effective approach for achieving in-vitro maturation of follicles. These techniques provide critical support for ovarian function in various ovarian-associated disorders, including primary ovarian insufficiency (POI), premature ovarian failure (POF), ovarian cancer, and age-related infertility.
Collapse
Affiliation(s)
- Negin Chavoshinezhad
- Immunology research center , Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomy, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behrooz Niknafs
- Immunology research center , Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Anatomy, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Chen C, Boché A, Wang Z, Lopez E, Peng J, Carreiras F, Schanne-Klein MC, Chen Y, Lambert A, Aimé C. The Balance Between Shear Flow and Extracellular Matrix in Ovarian Cancer-on-Chip. Adv Healthc Mater 2024; 13:e2400938. [PMID: 38829702 DOI: 10.1002/adhm.202400938] [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: 03/12/2024] [Revised: 04/30/2024] [Indexed: 06/05/2024]
Abstract
Ovarian cancer is the most lethal gynecologic cancer in developed countries. In the tumor microenvironment, the extracellular matrix (ECM) and flow shear stress are key players in directing ovarian cancer cells invasion. Artificial ECM models based only on ECM proteins are used to build an ovarian tumor-on-chip to decipher the crosstalk between ECM and shear stress on the migratory behavior and cellular heterogeneity of ovarian tumor cells. This work shows that in the shear stress regime of the peritoneal cavity, the ECM plays a major role in driving individual or collective ovarian tumor cells migration. In the presence of basement membrane proteins, migration is more collective than on type I collagen regardless of shear stress. With increasing shear stress, individual cell migration is enhanced; while, no significant impact on collective migration is measured. This highlights the central position that ECM and flow shear stress should hold in in vitro ovarian cancer models to deepen understanding of cellular responses and improve development of ovarian cancer therapeutic platforms. In this frame, adding flow provides significant improvement in biological relevance over the authors' previous work. Further steps for enhanced clinical relevance require not only multiple cell lines but also patient-derived cells and sera.
Collapse
Affiliation(s)
- Changchong Chen
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Alphonse Boché
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Groupe Matrice Extracellulaire et physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, Cergy, 95000, France
| | - Zixu Wang
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Elliot Lopez
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Juan Peng
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Franck Carreiras
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Groupe Matrice Extracellulaire et physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, Cergy, 95000, France
| | - Marie-Claire Schanne-Klein
- Laboratoire d'Optique et Biosciences (LOB), École polytechnique, CNRS, Inserm, Institut Polytechnique de Paris, Palaiseau, F-91128, France
| | - Yong Chen
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Ambroise Lambert
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Groupe Matrice Extracellulaire et physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, Cergy, 95000, France
| | - Carole Aimé
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| |
Collapse
|
17
|
Obaid G, Celli JP, Broekgaarden M, Bulin AL, Uusimaa P, Pogue B, Hasan T, Huang HC. Engineering photodynamics for treatment, priming and imaging. NATURE REVIEWS BIOENGINEERING 2024; 2:752-769. [PMID: 39927170 PMCID: PMC11801064 DOI: 10.1038/s44222-024-00196-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/07/2024] [Indexed: 02/11/2025]
Abstract
Photodynamic therapy (PDT) is a photochemistry-based treatment approach that relies on the activation of photosensitizers by light to locally generate reactive oxygen species that induce cellular cytotoxicity, in particular for the treatment of tumours. The cytotoxic effects of PDT are depth-limited owing to light penetration limits in tissue. However, photodynamic priming (PDP), which inherently occurs during PDT, can prime the tissue microenvironment to adjuvant therapies beyond the direct PDT ablative zone. In this Review, we discuss the underlying mechanisms of PDT and PDP, and their application to the treatment of cancer, outlining how PDP can permeabilize the tumour vasculature, overcome biological barriers, modulate multidrug resistance, enhance immune responses, increase tumour permeability and enable the photochemical release of drugs. We further examine the molecular engineering of photosensitizers to improve their pharmacodynamic and pharmacokinetic properties, increase their molecular specificity and allow image guidance of PDT, and investigate engineered cellular models for the design and optimization of PDT and PDP. Finally, we discuss alternative activation sources, including ultrasound, X-rays and self-illuminating compounds, and outline key barriers to the clinical translation of PDT and PDP.
Collapse
Affiliation(s)
- Girgis Obaid
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Jonathan P. Celli
- Department of Physics, University of Massachusetts Boston, Boston, MA, USA
| | - Mans Broekgaarden
- Grenoble Alpes University, INSERM U1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | - Anne-Laure Bulin
- Grenoble Alpes University, INSERM U1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | | | - Brian Pogue
- Department of Medical Physics, University of Wisconsin School of Medicine, Madison, WI, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| |
Collapse
|
18
|
Li W, Guo Z, Zhou Z, Zhou Z, He H, Sun J, Zhou X, Chin YR, Zhang L, Yang M. Distinguishing high-metastasis-potential circulating tumor cells through fluidic shear stress in a bloodstream-like microfluidic circulatory system. Oncogene 2024; 43:2295-2306. [PMID: 38858591 DOI: 10.1038/s41388-024-03075-4] [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: 12/22/2023] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024]
Abstract
Circulating tumor cells (CTCs) play a critical role as initiators in tumor metastasis, which unlocks an irreversible process of cancer progression. Regarding the fluid environment of intravascular CTCs, a comprehensive understanding of the impact of hemodynamic shear stress on CTCs is of profound significance but remains vague. Here, we report a microfluidic circulatory system that can emulate the CTC microenvironment to research the responses of typical liver cancer cells to varying levels of fluid shear stress (FSS). We observe that HepG2 cells surviving FSS exhibit a marked overexpression of TLR4 and TPPP3, which are shown to be associated with the colony formation, migration, and anti-apoptosis abilities of HepG2. Furthermore, overexpression of these two genes in another liver cancer cell line with normally low TLR4 and TPPP3 expression, SK-Hep-1 cells, by lentivirus-mediated transfection also confirms the critical role of TLR4 and TPPP3 in improving colony formation, migration, and survival capability under a fluid environment. Interestingly, in vivo experiments show SK-Hep-1 cells, overexpressed with these genes, have enhanced metastatic potential to the liver and lungs in mouse models via tail vein injection. Mechanistically, TLR4 and TPPP3 upregulated by FSS may increase FSS-mediated cell survival and metastasis through the p53-Bax signaling pathway. Moreover, elevated levels of these genes correlate with poorer overall survival in liver cancer patients, suggesting that our findings could offer new therapeutic strategies for early cancer diagnosis and targeted treatment development.
Collapse
Affiliation(s)
- Wenxiu Li
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Zhengjun Guo
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zhihang Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zhengdong Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Huimin He
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Jiayu Sun
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Y Rebecca Chin
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Liang Zhang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China.
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China.
| |
Collapse
|
19
|
Plesselova S, Calar K, Axemaker H, Sahly E, de la Puente P. Multicompartmentalized microvascularized tumor-on-a-chip to study tumor-stroma interactions and drug resistance in ovarian cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596456. [PMID: 38853974 PMCID: PMC11160770 DOI: 10.1101/2024.05.29.596456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
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.
Collapse
|
20
|
Gao W, Gu K, Ma L, Yang F, Deng L, Zhang Y, Miao MZ, Li W, Li G, Qian H, Zhang Z, Wang G, Yu H, Liu X. Interstitial Fluid Shear Stress Induces the Synthetic Phenotype Switching of VSMCs to Release Pro-calcified Extracellular Vesicles via EGFR-MAPK-KLF5 Pathway. Int J Biol Sci 2024; 20:2727-2747. [PMID: 38725857 PMCID: PMC11077359 DOI: 10.7150/ijbs.90725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 04/20/2024] [Indexed: 05/12/2024] Open
Abstract
Phenotypic switching (from contractile to synthetic) of vascular smooth muscle cells (VSMCs) is essential in the progression of atherosclerosis. The damaged endothelium in the atherosclerotic artery exposes VSMCs to increased interstitial fluid shear stress (IFSS). However, the precise mechanisms by which increased IFSS influences VSMCs phenotypic switching are unrevealed. Here, we employed advanced numerical simulations to calculate IFSS values accurately based on parameters acquired from patient samples. We then carefully investigated the phenotypic switching and extracellular vesicles (EVs) secretion of VSMCs under various IFSS conditions. By employing a comprehensive set of approaches, we found that VSMCs exhibited synthetic phenotype upon atherosclerotic IFSS. This synthetic phenotype is the upstream regulator for the enhanced secretion of pro-calcified EVs. Mechanistically, as a mechanotransducer, the epidermal growth factor receptor (EGFR) initiates the flow-based mechanical cues to MAPK signaling pathway, facilitating the nuclear accumulation of the transcription factor krüppel-like factor 5 (KLF5). Furthermore, pharmacological inhibiting either EGFR or MAPK signaling pathway blocks the nuclear accumulation of KLF5 and finally results in the maintenance of contractile VSMCs even under increased IFSS stimulation. Collectively, targeting this signaling pathway holds potential as a novel therapeutic strategy to inhibit VSMCs phenotypic switching and mitigate the progression of atherosclerosis.
Collapse
Affiliation(s)
- Wenbo Gao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Kaiyun Gu
- Department of Cardiac Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Lunjie Ma
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Fan Yang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Li Deng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yaojia Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Michael Z. Miao
- Division of Oral & Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Wenjun Li
- Division of Oral & Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Gang Li
- Department of Genome Sciences, University of Washington, William H. Foege Hall, 3720 15th Ave NE, Seattle 98195, USA
| | - Hong Qian
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Zhen Zhang
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
- JinFeng Laboratory, Chongqing 401329, China
| | - Hongchi Yu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
Gaebler D, Hachey SJ, Hughes CCW. Microphysiological systems as models for immunologically 'cold' tumors. Front Cell Dev Biol 2024; 12:1389012. [PMID: 38711620 PMCID: PMC11070549 DOI: 10.3389/fcell.2024.1389012] [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: 02/20/2024] [Accepted: 03/25/2024] [Indexed: 05/08/2024] Open
Abstract
The tumor microenvironment (TME) is a diverse milieu of cells including cancerous and non-cancerous cells such as fibroblasts, pericytes, endothelial cells and immune cells. The intricate cellular interactions within the TME hold a central role in shaping the dynamics of cancer progression, influencing pivotal aspects such as tumor initiation, growth, invasion, response to therapeutic interventions, and the emergence of drug resistance. In immunologically 'cold' tumors, the TME is marked by a scarcity of infiltrating immune cells, limited antigen presentation in the absence of potent immune-stimulating signals, and an abundance of immunosuppressive factors. While strategies targeting the TME as a therapeutic avenue in 'cold' tumors have emerged, there is a pressing need for novel approaches that faithfully replicate the complex cellular and non-cellular interactions in order to develop targeted therapies that can effectively stimulate immune responses and improve therapeutic outcomes in patients. Microfluidic devices offer distinct advantages over traditional in vitro 3D co-culture models and in vivo animal models, as they better recapitulate key characteristics of the TME and allow for precise, controlled insights into the dynamic interplay between various immune, stromal and cancerous cell types at any timepoint. This review aims to underscore the pivotal role of microfluidic systems in advancing our understanding of the TME and presents current microfluidic model systems that aim to dissect tumor-stromal, tumor-immune and immune-stromal cellular interactions in various 'cold' tumors. Understanding the intricacies of the TME in 'cold' tumors is crucial for devising effective targeted therapies to reinvigorate immune responses and overcome the challenges of current immunotherapy approaches.
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
|
22
|
Pajic-Lijakovic I, Milivojevic M, McClintock PVE. Role of viscoelasticity in the appearance of low-Reynolds turbulence: considerations for modelling. J Biol Eng 2024; 18:24. [PMID: 38589891 PMCID: PMC11476694 DOI: 10.1186/s13036-024-00415-6] [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: 01/08/2024] [Accepted: 02/24/2024] [Indexed: 04/10/2024] Open
Abstract
Inertial effects caused by perturbations of dynamical equilibrium during the flow of soft matter constitute a hallmark of turbulence. Such perturbations are attributable to an imbalance between energy storage and energy dissipation. During the flow of Newtonian fluids, kinetic energy can be both stored and dissipated, while the flow of viscoelastic soft matter systems, such as polymer fluids, induces the accumulation of both kinetic and elastic energies. The accumulation of elastic energy causes local stiffening of stretched polymer chains, which can destabilise the flow. Migrating multicellular systems are hugely complex and are capable of self-regulating their viscoelasticity and mechanical stress generation, as well as controlling their energy storage and energy dissipation. Since the flow perturbation of viscoelastic systems is caused by the inhomogeneous accumulation of elastic energy, rather than of kinetic energy, turbulence can occur at low Reynolds numbers.This theoretical review is focused on clarifying the role of viscoelasticity in the appearance of low-Reynolds turbulence. Three types of system are considered and compared: (1) high-Reynolds turbulent flow of Newtonian fluids, (2) low and moderate-Reynolds flow of polymer solutions, and (3) migration of epithelial collectives, discussed in terms of two model systems. The models considered involve the fusion of two epithelial aggregates, and the free expansion of epithelial monolayers on a substrate matrix.
Collapse
Affiliation(s)
- Ivana Pajic-Lijakovic
- Faculty of Technology and Metallurgy, Department of Chemical Engineering, University of Belgrade, Belgrade, Serbia.
| | - Milan Milivojevic
- Faculty of Technology and Metallurgy, Department of Chemical Engineering, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
23
|
Constantinescu DR, Sorop A, Ghionescu AV, Lixandru D, Herlea V, Bacalbasa N, Dima SO. EM-transcriptomic signature predicts drug response in advanced stages of high-grade serous ovarian carcinoma based on ascites-derived primary cultures. Front Pharmacol 2024; 15:1363142. [PMID: 38510654 PMCID: PMC10953505 DOI: 10.3389/fphar.2024.1363142] [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: 12/29/2023] [Accepted: 02/13/2024] [Indexed: 03/22/2024] Open
Abstract
Introduction: High-grade serous ovarian carcinoma (HGSOC) remains a medical challenge despite considerable improvements in the treatment. Unfortunately, over 75% of patients have already metastasized at the time of diagnosis. Advances in understanding the mechanisms underlying how ascites cause chemoresistance are urgently needed to derive novel therapeutic strategies. This study aimed to identify the molecular markers involved in drug sensitivity and highlight the use of ascites as a potential model to investigate HGSOC treatment options. Methods: After conducting an in silico analysis, eight epithelial-mesenchymal (EM)-associated genes related to chemoresistance were identified. To evaluate differences in EM-associated genes in HGSOC samples, we analyzed ascites-derived HGSOC primary cell culture (AS), tumor (T), and peritoneal nodule (NP) samples. Moreover, in vitro experiments were employed to measure tumor cell proliferation and cell migration in AS, following treatment with doxorubicin (DOX) and cisplatin (CIS) and expression of these markers. Results: Our results showed that AS exhibits a mesenchymal phenotype compared to tumor and peritoneal nodule samples. Moreover, DOX and CIS treatment leads to an invasive-intermediate epithelial-to-mesenchymal transition (EMT) state of the AS by different EM-associated marker expression. For instance, the treatment of AS showed that CDH1 and GATA6 decreased after CIS exposure and increased after DOX treatment. On the contrary, the expression of KRT18 has an opposite pattern. Conclusion: Taken together, our study reports a comprehensive investigation of the EM-associated genes after drug exposure of AS. Exploring ascites and their associated cellular and soluble components is promising for understanding the HGSOC progression and treatment response at a personalized level.
Collapse
Affiliation(s)
| | - Andrei Sorop
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
| | | | - Daniela Lixandru
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
- University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Vlad Herlea
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
- University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
- Department of Pathology-Fundeni Clinical Institute, Bucharest, Romania
| | - Nicolae Bacalbasa
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
- University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
- Center of Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Simona Olimpia Dima
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
- University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
- Center of Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| |
Collapse
|
24
|
Clevenger AJ, McFarlin MK, Gorley JPM, Solberg SC, Madyastha AK, Raghavan SA. Advances in cancer mechanobiology: Metastasis, mechanics, and materials. APL Bioeng 2024; 8:011502. [PMID: 38449522 PMCID: PMC10917464 DOI: 10.1063/5.0186042] [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: 11/03/2023] [Accepted: 02/12/2024] [Indexed: 03/08/2024] Open
Abstract
Within the tumor microenvironment (TME), tumor cells are exposed to numerous mechanical forces, both internally and externally, which contribute to the metastatic cascade. From the initial growth of the tumor to traveling through the vasculature and to the eventual colonization of distant organs, tumor cells are continuously interacting with their surroundings through physical contact and mechanical force application. The mechanical forces found in the TME can be simplified into three main categories: (i) shear stress, (ii) tension and strain, and (iii) solid stress and compression. Each force type can independently impact tumor growth and progression. Here, we review recent bioengineering strategies, which have been employed to establish the connection between mechanical forces and tumor progression. While many cancers are explored in this review, we place great emphasis on cancers that are understudied in their response to mechanical forces, such as ovarian and colorectal cancers. We discuss the major steps of metastatic transformation and present novel, recent advances in model systems used to study how mechanical forces impact the study of the metastatic cascade. We end by summarizing systems that incorporate multiple forces to expand the complexity of our understanding of how tumor cells sense and respond to mechanical forces in their environment. Future studies would also benefit from the inclusion of time or the aspect of mechanical memory to further enhance this field. While the knowledge of mechanical forces and tumor metastasis grows, developing novel materials and in vitro systems are essential to providing new insight into predicting, treating, and preventing cancer progression and metastasis.
Collapse
Affiliation(s)
| | - Maygan K. McFarlin
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA
| | - John Paul M. Gorley
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA
| | - Spencer C. Solberg
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA
| | - Anirudh K. Madyastha
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA
| | | |
Collapse
|
25
|
Yoon H, Sabaté Del Río J, Cho SW, Park TE. Recent advances in micro-physiological systems for investigating tumor metastasis and organotropism. LAB ON A CHIP 2024; 24:1351-1366. [PMID: 38303676 DOI: 10.1039/d3lc01033c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Tumor metastasis involves complex processes that traditional 2D cultures and animal models struggle to fully replicate. Metastatic tumors undergo a multitude of transformations, including genetic diversification, adaptation to diverse microenvironments, and modified drug responses, contributing significantly to cancer-related mortality. Micro-physiological systems (MPS) technology emerges as a promising approach to emulate the metastatic process by integrating critical biochemical, biomechanical, and geometrical cues at a microscale. These systems are particularly advantageous simulating metastasis organotropism, the phenomenon where tumors exhibit a preference for metastasizing to particular organs. Organotropism is influenced by various factors, such as tumor cell characteristics, unique organ microenvironments, and organ-specific vascular conditions, all of which can be effectively examined using MPS. This review surveys the recent developments in MPS research from the past five years, with a specific focus on their applications in replicating tumor metastasis and organotropism. Furthermore, we discuss the current limitations in MPS-based studies of organotropism and propose strategies for more accurately replicating and analyzing the intricate aspects of organ-specific metastasis, which is pivotal in the development of targeted therapeutic approaches against metastatic cancers.
Collapse
Affiliation(s)
- Heejeong Yoon
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| | - Jonathan Sabaté Del Río
- Center for Algorithmic and Robotized Synthesis (CARS), Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Seung Woo Cho
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| | - Tae-Eun Park
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| |
Collapse
|
26
|
Buckley M, Kramer M, Johnson B, Huskin G, Berry J, Sewell-Loftin MK. Mechanical activation and expression of HSP27 in epithelial ovarian cancer. Sci Rep 2024; 14:2856. [PMID: 38310132 PMCID: PMC10838328 DOI: 10.1038/s41598-024-52992-7] [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: 01/04/2023] [Accepted: 01/25/2024] [Indexed: 02/05/2024] Open
Abstract
Understanding the complex biomechanical tumor microenvironment (TME) is of critical importance in developing the next generation of anti-cancer treatment strategies. This is especially true in epithelial ovarian cancer (EOC), the deadliest of the gynecologic cancers due to recurrent disease or chemoresistance. However, current models of EOC progression provide little control or ability to monitor how changes in biomechanical parameters alter EOC cell behaviors. In this study, we present a microfluidic device designed to permit biomechanical investigations of the ovarian TME. Using this microtissue system, we describe how biomechanical stimulation in the form of tensile strains upregulate phosphorylation of HSP27, a heat shock protein implicated in ovarian cancer chemoresistance. Furthermore, EOC cells treated with strain demonstrate decreased response to paclitaxel in the in vitro vascularized TME model. The results provide a direct link to biomechanical regulation of HSP27 as a mediator of EOC chemoresistance, possibly explaining the failure of such therapies in some patients. The work presented here lays a foundation to elucidating mechanobiological regulation of EOC progression, including chemoresistance and could provide novel targets for anti-cancer therapeutics.
Collapse
Affiliation(s)
- Molly Buckley
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6thAvenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, UK
| | - Maranda Kramer
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6thAvenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, UK
| | - Bronte Johnson
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6thAvenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, UK
| | - Gillian Huskin
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6thAvenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, UK
| | - Joel Berry
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6thAvenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, UK
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, UK
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1824 6thAvenue South, Wallace Tumor Institute, Room 630A, Birmingham, AL, 35294, UK.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, UK.
| |
Collapse
|
27
|
Rahman Z, Bordoloi AD, Rouhana H, Tavasso M, van der Zon G, Garbin V, Ten Dijke P, Boukany PE. Interstitial flow potentiates TGF-β/Smad-signaling activity in lung cancer spheroids in a 3D-microfluidic chip. LAB ON A CHIP 2024; 24:422-433. [PMID: 38087979 PMCID: PMC10826459 DOI: 10.1039/d3lc00886j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/01/2023] [Indexed: 01/31/2024]
Abstract
Within the tumor microenvironment (TME), cancer cells use mechanotransduction pathways to convert biophysical forces to biochemical signals. However, the underlying mechanisms and functional significance of these pathways remain largely unclear. The upregulation of mechanosensitive pathways from biophysical forces such as interstitial flow (IF), leads to the activation of various cytokines, including transforming growth factor-β (TGF-β). TGF-β promotes in part via a Smad-dependent signaling pathway the epithelial-mesenchymal transition (EMT) in cancer cells. The latter process is linked to increased cancer cell motility and invasion. Current research models have limited ability to investigate the combined effects of biophysical forces (such as IF) and cytokines (TGF-β) in a 3D microenvironment. We used a 3D-matrix based microfluidic platform to demonstrate the potentiating effect of IF on exogenous TGF-β induced upregulation of the Smad-signaling activity and the expression of mesenchymal marker vimentin in A549 lung cancer spheroids. To monitor this, we used stably integrated fluorescent based reporters into the A549 cancer cell genome. Our results demonstrate that IF enhances exogenous TGF-β induced Smad-signaling activity in lung cancer spheroids embedded in a matrix microenvironment. In addition, we observed an increased cell motility for A549 spheroids when exposed to IF and TGF-β. Our 3D-microfluidic model integrated with real-time imaging provides a powerful tool for investigating cancer cell signaling and motility associated with invasion characteristics in a physiologically relevant TME.
Collapse
Affiliation(s)
- Zaid Rahman
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands.
| | - Ankur Deep Bordoloi
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands.
| | - Haifa Rouhana
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands.
| | - Margherita Tavasso
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands.
| | - Gerard van der Zon
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Valeria Garbin
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands.
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands.
| |
Collapse
|
28
|
Tadić V, Zhang W, Brozovic A. The high-grade serous ovarian cancer metastasis and chemoresistance in 3D models. Biochim Biophys Acta Rev Cancer 2024; 1879:189052. [PMID: 38097143 DOI: 10.1016/j.bbcan.2023.189052] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most frequent and aggressive type of epithelial ovarian cancer, with high recurrence rate and chemoresistance being the main issues in its clinical management. HGSOC is specifically challenging due to the metastatic dissemination via spheroids in the ascitic fluid. The HGSOC spheroids represent the invasive and chemoresistant cellular fraction, which is impossible to investigate in conventional two-dimensional (2D) monolayer cell cultures lacking critical cell-to-cell and cell-extracellular matrix interactions. Three-dimensional (3D) HGSOC cultures, where cells aggregate and exhibit relevant interactions, offer a promising in vitro model of peritoneal metastasis and multicellular drug resistance. This review summarizes recent studies of HGSOC in 3D culture conditions and highlights the role of multicellular HGSOC spheroids and ascitic environment in HGSOC metastasis and chemoresistance.
Collapse
Affiliation(s)
- Vanja Tadić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, Zagreb HR-10000, Croatia
| | - Wei Zhang
- Department of Engineering Mechanics, Dalian University of Technology, Linggong Road 2, Dalian CN-116024, China
| | - Anamaria Brozovic
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, Zagreb HR-10000, Croatia.
| |
Collapse
|
29
|
Yan J, Wu T, Zhang J, Gao Y, Wu JM, Wang S. Revolutionizing the female reproductive system research using microfluidic chip platform. J Nanobiotechnology 2023; 21:490. [PMID: 38111049 PMCID: PMC10729361 DOI: 10.1186/s12951-023-02258-7] [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: 08/19/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023] Open
Abstract
Comprehensively understanding the female reproductive system is crucial for safeguarding fertility and preventing diseases concerning women's health. With the capacity to simulate the intricate physio- and patho-conditions, and provide diagnostic platforms, microfluidic chips have fundamentally transformed the knowledge and management of female reproductive health, which will ultimately promote the development of more effective assisted reproductive technologies, treatments, and drug screening approaches. This review elucidates diverse microfluidic systems in mimicking the ovary, fallopian tube, uterus, placenta and cervix, and we delve into the culture of follicles and oocytes, gametes' manipulation, cryopreservation, and permeability especially. We investigate the role of microfluidics in endometriosis and hysteromyoma, and explore their applications in ovarian cancer, endometrial cancer and cervical cancer. At last, the current status of assisted reproductive technology and integrated microfluidic devices are introduced briefly. Through delineating the multifarious advantages and challenges of the microfluidic technology, we chart a definitive course for future research in the woman health field. As the microfluidic technology continues to evolve and advance, it holds great promise for revolutionizing the diagnosis and treatment of female reproductive health issues, thus propelling us into a future where we can ultimately optimize the overall wellbeing and health of women everywhere.
Collapse
Affiliation(s)
- Jinfeng Yan
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Tong Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Jinjin Zhang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Yueyue Gao
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Jia-Min Wu
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China.
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China.
| | - Shixuan Wang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
30
|
Murugan NJ, Cariba S, Abeygunawardena S, Rouleau N, Payne SL. Biophysical control of plasticity and patterning in regeneration and cancer. Cell Mol Life Sci 2023; 81:9. [PMID: 38099951 PMCID: PMC10724343 DOI: 10.1007/s00018-023-05054-6] [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: 08/18/2023] [Revised: 10/12/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023]
Abstract
Cells and tissues display a remarkable range of plasticity and tissue-patterning activities that are emergent of complex signaling dynamics within their microenvironments. These properties, which when operating normally guide embryogenesis and regeneration, become highly disordered in diseases such as cancer. While morphogens and other molecular factors help determine the shapes of tissues and their patterned cellular organization, the parallel contributions of biophysical control mechanisms must be considered to accurately predict and model important processes such as growth, maturation, injury, repair, and senescence. We now know that mechanical, optical, electric, and electromagnetic signals are integral to cellular plasticity and tissue patterning. Because biophysical modalities underly interactions between cells and their extracellular matrices, including cell cycle, metabolism, migration, and differentiation, their applications as tuning dials for regenerative and anti-cancer therapies are being rapidly exploited. Despite this, the importance of cellular communication through biophysical signaling remains disproportionately underrepresented in the literature. Here, we provide a review of biophysical signaling modalities and known mechanisms that initiate, modulate, or inhibit plasticity and tissue patterning in models of regeneration and cancer. We also discuss current approaches in biomedical engineering that harness biophysical control mechanisms to model, characterize, diagnose, and treat disease states.
Collapse
Affiliation(s)
- Nirosha J Murugan
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada.
- Allen Discovery Center, Tufts University, Medford, MA, USA.
| | - Solsa Cariba
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | | | - Nicolas Rouleau
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada
- Allen Discovery Center, Tufts University, Medford, MA, USA
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Samantha L Payne
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
31
|
Macaraniag C, Zhou J, Li J, Putzbach W, Hay N, Papautsky I. Microfluidic isolation of breast cancer circulating tumor cells from microvolumes of mouse blood. Electrophoresis 2023; 44:1859-1867. [PMID: 37528726 DOI: 10.1002/elps.202300108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 08/03/2023]
Abstract
Liquid biopsy has shown significant research and clinical implications in cancer. Particularly, the isolation of circulating tumor cells (CTCs) in preclinical studies can provide crucial information about disease progression and therefore may guide treatment decisions. Microfluidic isolation systems have played a considerable role in CTC isolation for cancer studies, disease diagnosis, and prognosis. CTCs are often studied using preclinical animal models such as xenografts or syngeneic models. However, most isolation systems are tested on human cell lines and human blood, whereas less validation studies are done on preclinical samples such as CTCs from mouse models. Here, we demonstrate and evaluate a complete workflow of a sized-based inertial microfluidic device to isolate CTCs from blood using exclusively mouse blood and mouse cancer cell lines. We then incorporate the cytospin, a commonly used method for enumeration of small number of cells in a glass slide to quantify the total cell yield of our workflow.
Collapse
Affiliation(s)
- Celine Macaraniag
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois, USA
| | - Jian Zhou
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois, USA
- University of Illinois Cancer Center, Chicago, Illinois, USA
| | - Jing Li
- Department of Biochemistry and Molecular Genetics, University of Illinois Chicago, Chicago, Illinois, USA
| | - William Putzbach
- Department of Biochemistry and Molecular Genetics, University of Illinois Chicago, Chicago, Illinois, USA
| | - Nissim Hay
- University of Illinois Cancer Center, Chicago, Illinois, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois Chicago, Chicago, Illinois, USA
| | - Ian Papautsky
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois, USA
- University of Illinois Cancer Center, Chicago, Illinois, USA
| |
Collapse
|
32
|
Xin Y, Li K, Huang M, Liang C, Siemann D, Wu L, Tan Y, Tang X. Biophysics in tumor growth and progression: from single mechano-sensitive molecules to mechanomedicine. Oncogene 2023; 42:3457-3490. [PMID: 37864030 PMCID: PMC10656290 DOI: 10.1038/s41388-023-02844-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.
Collapse
Grants
- R35 GM150812 NIGMS NIH HHS
- This work was financially supported by National Natural Science Foundation of China (Project no. 11972316, Y.T.), Shenzhen Science and Technology Innovation Commission (Project no. JCYJ20200109142001798, SGDX2020110309520303, and JCYJ20220531091002006, Y.T.), General Research Fund of Hong Kong Research Grant Council (PolyU 15214320, Y. T.), Health and Medical Research Fund (HMRF18191421, Y.T.), Hong Kong Polytechnic University (1-CD75, 1-ZE2M, and 1-ZVY1, Y.T.), the Cancer Pilot Research Award from UF Health Cancer Center (X. T.), the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM150812 (X. T.), the National Science Foundation under grant number 2308574 (X. T.), the Air Force Office of Scientific Research under award number FA9550-23-1-0393 (X. T.), the University Scholar Program (X. T.), UF Research Opportunity Seed Fund (X. T.), the Gatorade Award (X. T.), and the National Science Foundation REU Site at UF: Engineering for Healthcare (Douglas Spearot and Malisa Sarntinoranont). We are deeply grateful for the insightful discussions with and generous support from all members of Tang (UF)’s and Tan (PolyU)’s laboratories and all staff members of the MAE/BME/ECE/Health Cancer Center at UF and BME at PolyU.
- National Natural Science Foundation of China (National Science Foundation of China)
- Shenzhen Science and Technology Innovation Commission
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Chenyu Liang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar Siemann
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Lizi Wu
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
33
|
Espina JA, Cordeiro MH, Milivojevic M, Pajić-Lijaković I, Barriga EH. Response of cells and tissues to shear stress. J Cell Sci 2023; 136:jcs260985. [PMID: 37747423 PMCID: PMC10560560 DOI: 10.1242/jcs.260985] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023] Open
Abstract
Shear stress is essential for normal physiology and malignancy. Common physiological processes - such as blood flow, particle flow in the gut, or contact between migratory cell clusters and their substrate - produce shear stress that can have an impact on the behavior of different tissues. In addition, shear stress has roles in processes of biomedical interest, such as wound healing, cancer and fibrosis induced by soft implants. Thus, understanding how cells react and adapt to shear stress is important. In this Review, we discuss in vivo and in vitro data obtained from vascular and epithelial models; highlight the insights these have afforded regarding the general mechanisms through which cells sense, transduce and respond to shear stress at the cellular levels; and outline how the changes cells experience in response to shear stress impact tissue organization. Finally, we discuss the role of shear stress in collective cell migration, which is only starting to be appreciated. We review our current understanding of the effects of shear stress in the context of embryo development, cancer and fibrosis, and invite the scientific community to further investigate the role of shear stress in these scenarios.
Collapse
Affiliation(s)
- Jaime A. Espina
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), 2780-156 Oeiras, Portugal
| | - Marilia H. Cordeiro
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), 2780-156 Oeiras, Portugal
| | - Milan Milivojevic
- Faculty of Technology and Metallurgy, Belgrade University, 11120 Belgrade, Serbia
| | | | - Elias H. Barriga
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), 2780-156 Oeiras, Portugal
| |
Collapse
|
34
|
Gil JF, Moura CS, Silverio V, Gonçalves G, Santos HA. Cancer Models on Chip: Paving the Way to Large-Scale Trial Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300692. [PMID: 37103886 DOI: 10.1002/adma.202300692] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/05/2023] [Indexed: 06/19/2023]
Abstract
Cancer kills millions of individuals every year all over the world (Global Cancer Observatory). The physiological and biomechanical processes underlying the tumor are still poorly understood, hindering researchers from creating new, effective therapies. Inconsistent results of preclinical research, in vivo testing, and clinical trials decrease drug approval rates. 3D tumor-on-a-chip (ToC) models integrate biomaterials, tissue engineering, fabrication of microarchitectures, and sensory and actuation systems in a single device, enabling reliable studies in fundamental oncology and pharmacology. This review includes a critical discussion about their ability to reproduce the tumor microenvironment (TME), the advantages and drawbacks of existing tumor models and architectures, major components and fabrication techniques. The focus is on current materials and micro/nanofabrication techniques used to manufacture reliable and reproducible microfluidic ToC models for large-scale trial applications.
Collapse
Affiliation(s)
- João Ferreira Gil
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, Marinha Grande, 2430-028, Portugal
- INESC Microsistemas e Nanotecnologias (INESC MN), Rua Alves Redol 9, Lisbon, 1000-029, Portugal
- TEMA, Mechanical Engineering Department, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Carla Sofia Moura
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, Marinha Grande, 2430-028, Portugal
- Polytechnic Institute of Coimbra, Applied Research Institute, Coimbra, 3045-093, Portugal
| | - Vania Silverio
- INESC Microsistemas e Nanotecnologias (INESC MN), Rua Alves Redol 9, Lisbon, 1000-029, Portugal
- Department of Physics, Instituto Superior Técnico, Lisbon, 1049-001, Portugal
- Associate Laboratory Institute for Health and Bioeconomy - i4HB, Lisbon, Portugal
| | - Gil Gonçalves
- TEMA, Mechanical Engineering Department, University of Aveiro, Aveiro, 3810-193, Portugal
- Intelligent Systems Associate Laboratory (LASI), Aveiro, 3810-193, Portugal
| | - Hélder A Santos
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, 9713 AV, The Netherlands
- W.J. Korf Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| |
Collapse
|
35
|
Sorrin AJ, Zhou K, May K, Liu C, McNaughton K, Rahman I, Liang BJ, Rizvi I, Roque DM, Huang HC. Transient fluid flow improves photoimmunoconjugate delivery and photoimmunotherapy efficacy. iScience 2023; 26:107221. [PMID: 37520715 PMCID: PMC10372742 DOI: 10.1016/j.isci.2023.107221] [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: 11/15/2022] [Revised: 04/01/2023] [Accepted: 06/22/2023] [Indexed: 08/01/2023] Open
Abstract
Circulating drugs in the peritoneal cavity is an effective strategy for advanced ovarian cancer treatment. Photoimmunotherapy, an emerging modality with potential for the treatment of ovarian cancer, involves near-infrared light activation of antibody-photosensitizer conjugates (photoimmunoconjugates) to generate cytotoxic reactive oxygen species. Here, a microfluidic cell culture model is used to study how fluid flow-induced shear stress affects photoimmunoconjugate delivery to ovarian cancer cells. Photoimmunoconjugates are composed of the antibody, cetuximab, conjugated to the photosensitizer, and benzoporphyrin derivative. Longitudinal tracking of photoimmunoconjugate treatment under flow conditions reveals enhancements in subcellular photosensitizer accumulation. Compared to static conditions, fluid flow-induced shear stress at 0.5 and 1 dyn/cm2 doubled the cellular delivery of photoimmunoconjugates. Fluid flow-mediated treatment with three different photosensitizer formulations (benzoporphyrin derivative, photoimmunoconjugates, and photoimmunoconjugate-coated liposomes) led to enhanced phototoxicity compared to static conditions. This study confirms the fundamental role of fluid flow-induced shear stress in the anti-cancer effects of photoimmunotherapy.
Collapse
Affiliation(s)
- Aaron J. Sorrin
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Keri Zhou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Katherine May
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Cindy Liu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Kathryn McNaughton
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Idrisa Rahman
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Barry J. Liang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, North Carolina State University, Raleigh, NC 27599, USA
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dana M. Roque
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
36
|
Deng S, Li C, Cao J, Cui Z, Du J, Fu Z, Yang H, Chen P. Organ-on-a-chip meets artificial intelligence in drug evaluation. Theranostics 2023; 13:4526-4558. [PMID: 37649608 PMCID: PMC10465229 DOI: 10.7150/thno.87266] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/02/2023] [Indexed: 09/01/2023] Open
Abstract
Drug evaluation has always been an important area of research in the pharmaceutical industry. However, animal welfare protection and other shortcomings of traditional drug development models pose obstacles and challenges to drug evaluation. Organ-on-a-chip (OoC) technology, which simulates human organs on a chip of the physiological environment and functionality, and with high fidelity reproduction organ-level of physiology or pathophysiology, exhibits great promise for innovating the drug development pipeline. Meanwhile, the advancement in artificial intelligence (AI) provides more improvements for the design and data processing of OoCs. Here, we review the current progress that has been made to generate OoC platforms, and how human single and multi-OoCs have been used in applications, including drug testing, disease modeling, and personalized medicine. Moreover, we discuss issues facing the field, such as large data processing and reproducibility, and point to the integration of OoCs and AI in data analysis and automation, which is of great benefit in future drug evaluation. Finally, we look forward to the opportunities and challenges faced by the coupling of OoCs and AI. In summary, advancements in OoCs development, and future combinations with AI, will eventually break the current state of drug evaluation.
Collapse
Affiliation(s)
- Shiwen Deng
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Caifeng Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| | - Junxian Cao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhao Cui
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jiang Du
- Yunnan Biovalley Pharmaceutical Co., Ltd, Kunming 650503, China
| | - Zheng Fu
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| | - Hongjun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| | - Peng Chen
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Yunnan Biovalley Pharmaceutical Co., Ltd, Kunming 650503, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| |
Collapse
|
37
|
Zhou B, Feng Z, Xu J, Xie J. Organoids: approaches and utility in cancer research. Chin Med J (Engl) 2023; 136:1783-1793. [PMID: 37365679 PMCID: PMC10406116 DOI: 10.1097/cm9.0000000000002477] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Indexed: 06/28/2023] Open
Abstract
ABSTRACT Organoids are three-dimensional cellular structures with self-organizing and self-differentiation capacities. They faithfully recapitulate structures and functions of in vivo organs as represented by functionality and microstructural definitions. Heterogeneity in in vitro disease modeling is one of the main reasons for anti-cancer therapy failures. Establishing a powerful model to represent tumor heterogeneity is crucial for elucidating tumor biology and developing effective therapeutic strategies. Tumor organoids can retain the original tumor heterogeneity and are commonly used to mimic the cancer microenvironment when co-cultured with fibroblasts and immune cells; therefore, considerable effort has been made recently to promote the use of this new technology from basic research to clinical studies in tumors. In combination with gene editing technology and microfluidic chip systems, engineered tumor organoids show promising abilities to recapitulate tumorigenesis and metastasis. In many studies, the responses of tumor organoids to various drugs have shown a positive correlation with patient responses. Owing to these consistent responses and personalized characteristics with patient data, tumor organoids show excellent potential for preclinical research. Here, we summarize the properties of different tumor models and review their current state and progress in tumor organoids. We further discuss the substantial challenges and prospects in the rapidly developing tumor organoid field.
Collapse
Affiliation(s)
- Bingrui Zhou
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Zhiwei Feng
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jun Xu
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplant Center, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| |
Collapse
|
38
|
Li C, Holman JB, Shi Z, Qiu B, Ding W. On-chip modeling of tumor evolution: Advances, challenges and opportunities. Mater Today Bio 2023; 21:100724. [PMID: 37483380 PMCID: PMC10359640 DOI: 10.1016/j.mtbio.2023.100724] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
Tumor evolution is the accumulation of various tumor cell behaviors from tumorigenesis to tumor metastasis and is regulated by the tumor microenvironment (TME). However, the mechanism of solid tumor progression has not been completely elucidated, and thus, the development of tumor therapy is still limited. Recently, Tumor chips constructed by culturing tumor cells and stromal cells on microfluidic chips have demonstrated great potential in modeling solid tumors and visualizing tumor cell behaviors to exploit tumor progression. Herein, we review the methods of developing engineered solid tumors on microfluidic chips in terms of tumor types, cell resources and patterns, the extracellular matrix and the components of the TME, and summarize the recent advances of microfluidic chips in demonstrating tumor cell behaviors, including proliferation, epithelial-to-mesenchymal transition, migration, intravasation, extravasation and immune escape of tumor cells. We also outline the combination of tumor organoids and microfluidic chips to elaborate tumor organoid-on-a-chip platforms, as well as the practical limitations that must be overcome.
Collapse
Affiliation(s)
- Chengpan Li
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Joseph Benjamin Holman
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Zhengdi Shi
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Bensheng Qiu
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
- Center for Biomedical Imaging, 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
| |
Collapse
|
39
|
Li Q, Xiao X, Feng J, Yan R, Xi J. Machine learning-assisted analysis of epithelial mesenchymal transition pathway for prognostic stratification and immune infiltration assessment in ovarian cancer. Front Endocrinol (Lausanne) 2023; 14:1196094. [PMID: 37404304 PMCID: PMC10317337 DOI: 10.3389/fendo.2023.1196094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/24/2023] [Indexed: 07/06/2023] Open
Abstract
Background Ovarian cancer is the most lethal gynaecological malignancy, and serous ovarian cancer (SOC) is one of the more important pathological subtypes. Previous studies have reported a significant association of epithelial tomesenchymal transition (EMT) with invasive metastasis and immune modulation of SOC, however, there is a lack of prognostic and immune infiltration biomarkers reported for SOC based on EMT. Methods Gene expression data for ovarian cancer and corresponding patient clinical data were collected from the TCGA database and the GEO database, and cell type annotation and spatial expression analysis were performed on single cell sequencing data from the GEO database. To understand the cell type distribution of EMT-related genes in SOC single-cell data and the enrichment relationships of biological pathways and tumour functions. In addition, GO functional annotation analysis and KEGG pathway enrichment analysis were performed on mRNAs predominantly expressed with EMT to predict the biological function of EMT in ovarian cancer. The major differential genes of EMT were screened to construct a prognostic risk prediction model for SOC patients. Data from 173 SOC patient samples obtained from the GSE53963 database were used to validate the prognostic risk prediction model for ovarian cancer. Here we also analysed the direct association between SOC immune infiltration and immune cell modulation and EMT risk score. and calculate drug sensitivity scores in the GDSC database.In addition, we assessed the specific relationship between GAS1 gene and SOC cell lines. Results Single cell transcriptome analysis in the GEO database annotated the major cell types of SOC samples, including: T cell, Myeloid, Epithelial cell, Fibroblast, Endothelial cell, and Bcell. cellchat revealed several cell type interactions that were shown to be associated with EMT-mediated SOC invasion and metastasis. A prognostic stratification model for SOC was constructed based on EMT-related differential genes, and the Kapan-Meier test showed that this biomarker had significant prognostic stratification value for several independent SOC databases. The EMT risk score has good stratification and identification properties for drug sensitivity in the GDSC database. Conclusions This study constructed a prognostic stratification biomarker based on EMT-related risk genes for immune infiltration mechanisms and drug sensitivity analysis studies in SOC. This lays the foundation for in-depth clinical studies on the role of EMT in immune regulation and related pathway alterations in SOC. It is also hoped to provide effective potential solutions for early diagnosis and clinical treatment of ovarian cancer.
Collapse
|
40
|
Melnik D, Cortés-Sánchez JL, Sandt V, Kahlert S, Kopp S, Grimm D, Krüger M. Dexamethasone Selectively Inhibits Detachment of Metastatic Thyroid Cancer Cells during Random Positioning. Cancers (Basel) 2023; 15:cancers15061641. [PMID: 36980530 PMCID: PMC10046141 DOI: 10.3390/cancers15061641] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
We recently reported that synthetic glucocorticoid dexamethasone (DEX) is able to suppress metastasis-like spheroid formation in a culture of follicular thyroid cancer (FTC)-133 cells cultured under random positioning. We now show that this inhibition was selective for two metastatic thyroid carcinoma cells, FTC-133 and WRO, whereas benign Nthy-ori 3-1 thyrocytes and recurrent ML-1 follicular thyroid cancer cells were not affected by DEX. We then compare Nthy-ori 3-1 and FTC-133 cells concerning their adhesion and mechanosignaling. We demonstrate that DEX disrupts random positioning-triggered p38 stress signaling in FTC-133 cells, thereby antagonizing a variety of biological functions. Thus, DEX treatment of FTC-133 cells is associated with increased adhesiveness, which is mainly caused by the restored, pronounced formation of a normal number of tight junctions. Moreover, we show that Nthy-ori 3-1 and ML-1 cells upregulate the anti-adhesion protein mucin-1 during random positioning, presumably as a protection against mechanical stress. In summary, mechanical stress seems to be an important component in this metastasis model system that is processed differently by metastatic and healthy cells. The balance between adhesion, anti-adhesion and cell–cell connections enables detachment of adherent human cells on the random positioning machine—or not, allowing selective inhibition of thyroid in vitro metastasis by DEX.
Collapse
Affiliation(s)
- Daniela Melnik
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - José Luis Cortés-Sánchez
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
| | - Viviann Sandt
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
| | - Stefan Kahlert
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Institute of Anatomy, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Sascha Kopp
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Core Facility Tissue Engineering, Otto von Guericke University, 39106 Magdeburg, Germany
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Correspondence: ; Tel.: +49-391-6757471
| |
Collapse
|
41
|
Rickard BP, Overchuk M, Obaid G, Ruhi MK, Demirci U, Fenton SE, Santos JH, Kessel D, Rizvi I. Photochemical Targeting of Mitochondria to Overcome Chemoresistance in Ovarian Cancer †. Photochem Photobiol 2023; 99:448-468. [PMID: 36117466 PMCID: PMC10043796 DOI: 10.1111/php.13723] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022]
Abstract
Ovarian cancer is the most lethal gynecologic malignancy with a stubborn mortality rate of ~65%. The persistent failure of multiline chemotherapy, and significant tumor heterogeneity, has made it challenging to improve outcomes. A target of increasing interest is the mitochondrion because of its essential role in critical cellular functions, and the significance of metabolic adaptation in chemoresistance. This review describes mitochondrial processes, including metabolic reprogramming, mitochondrial transfer and mitochondrial dynamics in ovarian cancer progression and chemoresistance. The effect of malignant ascites, or excess peritoneal fluid, on mitochondrial function is discussed. The role of photodynamic therapy (PDT) in overcoming mitochondria-mediated resistance is presented. PDT, a photochemistry-based modality, involves the light-based activation of a photosensitizer leading to the production of short-lived reactive molecular species and spatiotemporally confined photodamage to nearby organelles and biological targets. The consequential effects range from subcytotoxic priming of target cells for increased sensitivity to subsequent treatments, such as chemotherapy, to direct cell killing. This review discusses how PDT-based approaches can address key limitations of current treatments. Specifically, an overview of the mechanisms by which PDT alters mitochondrial function, and a summary of preclinical advancements and clinical PDT experience in ovarian cancer are provided.
Collapse
Affiliation(s)
- Brittany P. Rickard
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marta Overchuk
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; North Carolina State University, Raleigh, NC 27606, USA
| | - Girgis Obaid
- Department of Bioengineering, University of Texas at Dallas, Richardson TX 95080, USA
| | - Mustafa Kemal Ruhi
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Suzanne E. Fenton
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Mechanistic Toxicology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Janine H. Santos
- Mechanistic Toxicology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - David Kessel
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Imran Rizvi
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; North Carolina State University, Raleigh, NC 27606, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
42
|
Ngo H, Amartumur S, Tran VTA, Tran M, Diep YN, Cho H, Lee LP. In Vitro Tumor Models on Chip and Integrated Microphysiological Analysis Platform (MAP) for Life Sciences and High-Throughput Drug Screening. BIOSENSORS 2023; 13:231. [PMID: 36831997 PMCID: PMC9954135 DOI: 10.3390/bios13020231] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/23/2023] [Accepted: 01/31/2023] [Indexed: 06/18/2023]
Abstract
The evolution of preclinical in vitro cancer models has led to the emergence of human cancer-on-chip or microphysiological analysis platforms (MAPs). Although it has numerous advantages compared to other models, cancer-on-chip technology still faces several challenges such as the complexity of the tumor microenvironment and integrating multiple organs to be widely accepted in cancer research and therapeutics. In this review, we highlight the advancements in cancer-on-chip technology in recapitulating the vital biological features of various cancer types and their applications in life sciences and high-throughput drug screening. We present advances in reconstituting the tumor microenvironment and modeling cancer stages in breast, brain, and other types of cancer. We also discuss the relevance of MAPs in cancer modeling and precision medicine such as effect of flow on cancer growth and the short culture period compared to clinics. The advanced MAPs provide high-throughput platforms with integrated biosensors to monitor real-time cellular responses applied in drug development. We envision that the integrated cancer MAPs has a promising future with regard to cancer research, including cancer biology, drug discovery, and personalized medicine.
Collapse
Affiliation(s)
- Huyen Ngo
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sarnai Amartumur
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Van Thi Ai Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Minh Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yen N. Diep
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Luke P. Lee
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Bioengineering, University of California at Berkeley, Berkeley, CA 94720, USA
- Department of Electrical Engineering and Computer Science, University of California at Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
43
|
Pajic-Lijakovic I, Eftimie R, Milivojevic M, Bordas SPA. The dynamics along the biointerface between the epithelial and cancer mesenchymal cells: Modeling consideration. Semin Cell Dev Biol 2023; 147:47-57. [PMID: 36631334 DOI: 10.1016/j.semcdb.2022.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023]
Abstract
Epithelial cancer is the one of most lethal cancer type worldwide. Targeting the early stage of disease would allow dramatic improvements in the survival of cancer patients. The early stage of the disease is related to cancer cell spreading across surrounding healthy epithelium. Consequently, deeper insight into cell dynamics along the biointerface between epithelial and cancer (mesenchymal) cells is necessary in order to control the disease as soon as possible. Cell dynamics along this epithelial-cancer biointerface is the result of the interplay between various biological and physical mechanisms. Despite extensive research devoted to study cancer cell spreading across the epithelium, we still do not understand the physical mechanisms which influences the dynamics along the biointerface. These physical mechanisms are related to the interplay between physical parameters such as: (1) interfacial tension between cancer and epithelial subpopulations, (2) established interfacial tension gradients, (3) the bending rigidity of the biointerface and its impact on the interfacial tension, (4) surface tension of the subpopulations, (5) viscoelasticity caused by collective cell migration, and (6) cell residual stress accumulation. The main goal of this study is to review some of these physical parameters in the context of the epithelial/cancer biointerface elaborated on the model system such as the biointerface between breast epithelial MCF-10A cells and cancer MDA-MB-231 cells and then to incorporate these parameters into a new biophysical model that could describe the dynamics of the biointerface. We conclude by discussing three biophysical scenarios for cell dynamics along the biointerface, which can occur depending on the magnitude of the generated shear stress: a smooth biointerface, a slightly-perturbed biointerface and an intensively-perturbed biointerface in the context of the Kelvin-Helmholtz instability. These scenarios are related to the probability of cancer invasion.
Collapse
Affiliation(s)
- Ivana Pajic-Lijakovic
- University of Belgrade, Faculty of Technology and Metallurgy, Department of Chemical Engineering, Serbia.
| | - Raluca Eftimie
- Laboratoire Mathematiques de Besançon, UMR-CNRS 6623, Université de Bourgogne Franche-Comte, 16 Route de Gray, Besançon 25000, France
| | - Milan Milivojevic
- University of Belgrade, Faculty of Technology and Metallurgy, Department of Chemical Engineering, Serbia
| | - Stéphane P A Bordas
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan; Institute for Computational Engineering, Faculty of Science, Technology and Communication, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
44
|
Patient-Derived In Vitro Models of Ovarian Cancer: Powerful Tools to Explore the Biology of the Disease and Develop Personalized Treatments. Cancers (Basel) 2023; 15:cancers15020368. [PMID: 36672318 PMCID: PMC9856518 DOI: 10.3390/cancers15020368] [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: 12/02/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Epithelial ovarian cancer (OC) is the most lethal gynecological malignancy worldwide due to a late diagnosis caused by the lack of specific symptoms and rapid dissemination into the peritoneal cavity. The standard of care for OC treatment is surgical cytoreduction followed by platinum-based chemotherapy. While a response to this frontline treatment is common, most patients undergo relapse within 2 years and frequently develop a chemoresistant disease that has become unresponsive to standard treatments. Moreover, also due to the lack of actionable mutations, very few alternative therapeutic strategies have been designed as yet for the treatment of recurrent OC. This dismal clinical perspective raises the need for pre-clinical models that faithfully recapitulate the original disease and therefore offer suitable tools to design novel therapeutic approaches. In this regard, patient-derived models are endowed with high translational relevance, as they can better capture specific aspects of OC such as (i) the high inter- and intra-tumor heterogeneity, (ii) the role of cancer stem cells (a small subset of tumor cells endowed with tumor-initiating ability, which can sustain tumor spreading, recurrence and chemoresistance), and (iii) the involvement of the tumor microenvironment, which interacts with tumor cells and modulates their behavior. This review describes the different in vitro patient-derived models that have been developed in recent years in the field of OC research, focusing on their ability to recapitulate specific features of this disease. We also discuss the possibilities of leveraging such models as personalized platforms to design new therapeutic approaches and guide clinical decisions.
Collapse
|
45
|
Lopez E, Kamboj S, Chen C, Wang Z, Kellouche S, Leroy-Dudal J, Carreiras F, Lambert A, Aimé C. In Vitro Models of Ovarian Cancer: Bridging the Gap between Pathophysiology and Mechanistic Models. Biomolecules 2023; 13:biom13010103. [PMID: 36671488 PMCID: PMC9855568 DOI: 10.3390/biom13010103] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 01/06/2023] Open
Abstract
Ovarian cancer (OC) is a disease of major concern with a survival rate of about 40% at five years. This is attributed to the lack of visible and reliable symptoms during the onset of the disease, which leads over 80% of patients to be diagnosed at advanced stages. This implies that metastatic activity has advanced to the peritoneal cavity. It is associated with both genetic and phenotypic heterogeneity, which considerably increase the risks of relapse and reduce the survival rate. To understand ovarian cancer pathophysiology and strengthen the ability for drug screening, further development of relevant in vitro models that recapitulate the complexity of OC microenvironment and dynamics of OC cell population is required. In this line, the recent advances of tridimensional (3D) cell culture and microfluidics have allowed the development of highly innovative models that could bridge the gap between pathophysiology and mechanistic models for clinical research. This review first describes the pathophysiology of OC before detailing the engineering strategies developed to recapitulate those main biological features.
Collapse
Affiliation(s)
- Elliot Lopez
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Sahil Kamboj
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe, EA1391, Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, CEDEX, 95031 Neuville sur Oise, France
| | - Changchong Chen
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Zixu Wang
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Sabrina Kellouche
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe, EA1391, Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, CEDEX, 95031 Neuville sur Oise, France
| | - Johanne Leroy-Dudal
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe, EA1391, Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, CEDEX, 95031 Neuville sur Oise, France
| | - Franck Carreiras
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe, EA1391, Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, CEDEX, 95031 Neuville sur Oise, France
| | - Ambroise Lambert
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe, EA1391, Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, CEDEX, 95031 Neuville sur Oise, France
| | - Carole Aimé
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
- Correspondence:
| |
Collapse
|
46
|
Jin Y, Cai W, Zhao C, Yang F, Yang C, Zhang X, Zhou Q, Zhao W, Zhang C, Zhang F, Wang M, Li M. EMT status of circulating breast cancer cells and impact of fluidic shear stress. Exp Cell Res 2022; 421:113385. [PMID: 36228736 DOI: 10.1016/j.yexcr.2022.113385] [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: 07/05/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 12/29/2022]
Abstract
Circulating tumor cells (CTCs) play a vital role in the metastasis and recurrence of breast cancer. CTCs are highly heterogeneous at the stage of Epithelial-to-Mesenchymal Transition (EMT), but the phenotypic and biological characteristics in different EMT stages remain poorly defined. We conducted an orthotopic mouse (4T1) model of breast cancer to isolate CTCs and identified two phenotypes of CTCs: intermediate E/M and mesenchymal CTCs. MTT, Colony formation, Transwell migration and invasion assays were utilized to examined cell proliferation, colony forming, migration and invasion ability. Both the intermediate E/M and mesenchymal CTCs exhibited lower rates of proliferation, colony formation and invasion, as compared to primary tumor cells. The mesenchymal CTCs had a higher rate of invasion but lower rates of proliferation and colony formation than the intermediate E/M CTCs. They also exhibited lower rates of growth and metastasis than the primary tumor cells in vivo, but the mesenchymal CTCs had a higher rate of metastasis than the intermediate E/M CTCs. Fluid shear stress induced the EMT transition of CTCs. The comprehensive analysis of CTCs proteomics discovered proteins that differentially expressed in the two types of CTCs and their primary tumor cells.
Collapse
Affiliation(s)
- Yanling Jin
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Wei Cai
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Gansu Provincial Hospital, Lanzhou, China
| | - Chanyuan Zhao
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Feng Yang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Chenguang Yang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaoyu Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Quan Zhou
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Wenjie Zhao
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Chenli Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Fangfang Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Min Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Experimental Teaching Center of Basic Medicine, School of Basic Medical Science, Lanzhou University, Lanzhou, China.
| | - Min Li
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Gansu Provincial Key Laboratory of Preclinical Study for New Drug Development, Lanzhou University, Lanzhou, China.
| |
Collapse
|
47
|
Scemama A, Lunetto S, Biddle A. Highlight: microfluidic devices for cancer metastasis studies. IN VITRO MODELS 2022; 1:399-403. [PMID: 39872614 PMCID: PMC11756437 DOI: 10.1007/s44164-022-00023-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 01/30/2025]
Abstract
Whilst cancer is the single most researched disease by number of clinical trials, it remains a leading cause of death in middle- to high-income countries, with metastasis as the prime factor in cancer-associated mortality. The complex, multistep process of metastasis, in which cancer cells disseminate from the primary tumour and home to distant sites, is modulated by an extensive set of factors such as matrix properties, cancer cell plasticity, intercellular communication and oxygen tension. However, traditional treatment approaches have been focussed on the removal of the primary and secondary lesions, rather than interference with the metastatic cascade. Microfluidic platforms enable the deconvolution of the processes involved in metastasis by recapitulating key aspects of the tumour microenvironment in a controlled and reproducible fashion. Herein, we review recent developments in microfluidics for metastasis research and explain how these devices offer exceptional potential towards gaining a deeper understanding of this key aspect of malignancy.
Collapse
Affiliation(s)
- Alice Scemama
- Blizard Institute, Queen Mary University of London, London, UK
| | - Sophia Lunetto
- Blizard Institute, Queen Mary University of London, London, UK
| | - Adrian Biddle
- Blizard Institute, Queen Mary University of London, London, UK
| |
Collapse
|
48
|
Velasco V, Soucy P, Keynton R, Williams SJ. A microfluidic impedance platform for real-time, in vitro characterization of endothelial cells undergoing fluid shear stress. LAB ON A CHIP 2022; 22:4705-4716. [PMID: 36349980 DOI: 10.1039/d2lc00555g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
We introduce a microfluidic impedance platform to electrically monitor in real-time, endothelium monolayers undergoing fluid shear stress. Our platform incorporates sensing electrodes (SEs) that measure cell behavior and cell-free control electrodes that measure cell culture media resistance simultaneously but independently from SEs. We evaluated three different cellular subpopulations sizes through 50, 100, and 200 μm diameter SEs. We tested their utility in measuring the response of human umbilical vein endothelial cells (HUVECs) at static, constant (17.6 dyne per cm2), and stepped (23.7-35-58.1 dyne per cm2) shear stress conditions. For 14 hours, we collected the impedance spectra (100 Hz-1 MHz) of sheared cells. Using equivalent circuit models, we extracted monolayer permeability (RTER), cell membrane capacitance, and cell culture media resistance. Platform evaluation concluded that: (1) 50 μm SEs (∼2 cells) suffered interfacial capacitance and reduced cell measurement sensitivity, (2) 100 μm SEs (∼6 cells) was limited to measuring cell behavior only and cannot measure cell culture media resistance, and (3) 200 μm SEs (∼20 cells) detected cell behavior with accurate prediction of cell culture media resistance. Platform-based shear stress studies indicated a shear magnitude dependent increase in RTER at the onset of acute flow. Consecutive stepped shear conditions did not alter RTER in the same magnitude after shear has been applied. Finally, endpoint staining of VE-cadherin on the actual SEs and endpoint RTER measurements were greater for 23.7-35-58.1 dyne per cm2 than 17.6 dyne per cm2 shear conditions.
Collapse
Affiliation(s)
- Vanessa Velasco
- Stanford Genome Technology Center, Stanford University, 3165 Porter Drive, Palo Alto, CA 94304, USA
| | - Patricia Soucy
- Bioengineering Department, University of Louisville, 2301 S. Third St., Paul C. Lutz Hall, # 419, Louisville, KY 40292, USA
| | - Robert Keynton
- William States Lee College of Engineering, University of North Carolina, Charlotte, 28223, USA
| | - Stuart J Williams
- Mechanical Engineering Department, University of Louisville, 332 Eastern Pkwy, Sackett Hall, # 202A, Louisville, KY 40292, USA
| |
Collapse
|
49
|
Nguyen LTS, Jacob MAC, Parajón E, Robinson DN. Cancer as a biophysical disease: Targeting the mechanical-adaptability program. Biophys J 2022; 121:3573-3585. [PMID: 35505610 PMCID: PMC9617128 DOI: 10.1016/j.bpj.2022.04.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/30/2022] [Accepted: 04/28/2022] [Indexed: 11/02/2022] Open
Abstract
With the number of cancer cases projected to significantly increase over time, researchers are currently exploring "nontraditional" research fields in the pursuit of novel therapeutics. One emerging area that is steadily gathering interest revolves around cellular mechanical machinery. When looking broadly at the physical properties of cancer, it has been debated whether a cancer could be defined as either stiffer or softer across cancer types. With numerous articles supporting both sides, the evidence instead suggests that cancer is not particularly regimented. Instead, cancer is highly adaptable, allowing it to endure the constantly changing microenvironments cancer cells encounter, such as tumor compression and the shear forces in the vascular system and body. What allows cancer cells to achieve this adaptability are the particular proteins that make up the mechanical network, leading to a particular mechanical program of the cancer cell. Coincidentally, some of these proteins, such as myosin II, α-actinins, filamins, and actin, have either altered expression in cancer and/or some type of direct involvement in cancer progression. For this reason, targeting the mechanical system as a therapeutic strategy may lead to more efficacious treatments in the future. However, targeting the mechanical program is far from trivial. As involved as the mechanical program is in cancer development and metastasis, it also helps drive many other key cellular processes, such as cell division, cell adhesion, metabolism, and motility. Therefore, anti-cancer treatments targeting the mechanical program must take great care to avoid potential side effects. Here, we introduce the potential of targeting the mechanical program while also providing its challenges and shortcomings as a strategy for cancer treatment.
Collapse
Affiliation(s)
- Ly T S Nguyen
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Mark Allan C Jacob
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Eleana Parajón
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Douglas N Robinson
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
50
|
Macaraniag C, Luan Q, Zhou J, Papautsky I. Microfluidic techniques for isolation, formation, and characterization of circulating tumor cells and clusters. APL Bioeng 2022; 6:031501. [PMID: 35856010 PMCID: PMC9288269 DOI: 10.1063/5.0093806] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/28/2022] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cell (CTC) clusters that are shed from the primary tumor into the bloodstream are associated with a poor prognosis, elevated metastatic potential, higher proliferation rate, and distinct molecular features compared to single CTCs. Studying CTC clusters may give us information on the differences in the genetic profiles, somatic mutations, and epigenetic changes in circulating cells compared to the primary tumor and metastatic sites. Microfluidic systems offer the means of studying CTC clusters through the ability to efficiently isolate these rare cells from the whole blood of patients in a liquid biopsy. Microfluidics can also be used to develop in vitro models of CTC clusters and make possible their characterization and analysis. Ultimately, microfluidic systems can offer the means to gather insight on the complexities of the metastatic process, the biology of cancer, and the potential for developing novel or personalized therapies. In this review, we aim to discuss the advantages and challenges of the existing microfluidic systems for working with CTC clusters. We hope that an improved understanding of the role microfluidics can play in isolation, formation, and characterization of CTC clusters, which can lead to increased sophistication of microfluidic platforms in cancer research.
Collapse
Affiliation(s)
- Celine Macaraniag
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Qiyue Luan
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Jian Zhou
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Ian Papautsky
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|