1
|
de la Jara Ortiz F, Cimmino C, Ventre M, Cambi A. Understanding and measuring mechanical signals in the tumor stroma. FEBS Open Bio 2024. [PMID: 39523476 DOI: 10.1002/2211-5463.13923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/30/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
The tumor microenvironment (TME) is well known for its immune suppressive role, especially in solid tumors which are characterized by a thick, dense stroma. Apart from cell-cell interactions and biochemical signals, the tumor stroma is also characterized by its distinct mechanical properties, which are dictated by the composition and architecture of its extracellular matrix (ECM). Cancer-associated fibroblasts (CAFs) are the main producers and remodelers of the stromal ECM, and their heterogeneity has recently become a focus of intense research. This review describes recent findings highlighting CAF subtypes and their specific functions, as well as the development of 3D models to study tumor stroma mechanics in vitro. Finally, we discuss the quantitative techniques used to measure tissue mechanical properties at different scales. Given the diagnostic and prognostic value of stroma stiffness and composition, and the recent development of anti-tumor therapeutic strategies targeting the stroma, understanding and measuring tumor stroma mechanical properties has never been more timely or relevant.
Collapse
Affiliation(s)
- Fàtima de la Jara Ortiz
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Chiara Cimmino
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for Healthcare@CRIB, Fondazione Istituto Italiano di Tecnologia, Naples, Italy
| | - Maurizio Ventre
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for Healthcare@CRIB, Fondazione Istituto Italiano di Tecnologia, Naples, Italy
- Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Naples, Italy
| | - Alessandra Cambi
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
2
|
Zhong L, Li B, Zhang S, Li Q, Xiao G. Computational Identification of Migrating T cells in Spatial Transcriptomics Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619870. [PMID: 39484480 PMCID: PMC11526994 DOI: 10.1101/2024.10.23.619870] [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
T cells are the central players in antitumor immunity, and effective tumor killing depends on their ability to infiltrate into the tumor microenvironment (TME) while maintaining normal cytotoxicity. However, late-stage tumors develop immunosuppressive mechanisms that impede T cell movement and induce exhaustion. Investigating T cell migration in human tumors in vivo could provide novel insights into tumor immune escape, although it remains a challenging task. In this study, we developed ReMiTT, a computational method that leverages spatial transcriptomics data to track T cell migration patterns within tumor tissue. Applying ReMiTT to ovarian tumor samples, we identified key genes and pathways that enriched on algorithm-identified T cell migration trails, including leukocyte chemotaxis, cell-cell adhesion, and ECM remodeling. We also characterized the phenotypes of T cells on the migrating trails, suggesting that regulatory T cells may accompany cytotoxic T cells during migration. Our findings provide a novel approach to studying T cell migration and interactions within the TME, offering new insights into tumor-immune dynamics.
Collapse
|
3
|
Tolentino MAK, Seyedzadeh MH, Peres NG, Du EY, Zhu L, Gaus K, Goyette J, Gooding JJ. Polyethylene Glycol-Based Hydrogel as a 3D Extracellular Matrix Mimic for Cytotoxic T Lymphocytes. J Biomed Mater Res A 2024. [PMID: 39429059 DOI: 10.1002/jbm.a.37811] [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: 07/30/2024] [Revised: 09/09/2024] [Accepted: 10/05/2024] [Indexed: 10/22/2024]
Abstract
Three-dimensional (3D) in vitro models enable us to understand cell behavior that is a better reflection of what occurs in vivo than 2D in vitro models. As a result, developing 3D models for extracellular matrix (ECM) has been growing exponentially. Most of the efforts for these 3D models are geared toward understanding cancer cells. An intricate network of cells that engages with cancer cells and can kill them are the immune cells, particularly cytotoxic T lymphocytes (CTLs). However, limited reports are available for 3D ECM mimics to understand CTL dynamics. Currently, we lack ECM mimetic hydrogels for immune cells, with sufficient control over variables, such as stiffness, to fully understand CTL dynamics in vitro. Here, we developed PEG-based hydrogels as ECM mimics for CTLs. The ECM mimics are targeted to mimic the stiffness of soft tissues where CTLs reside, migrate, and deliver their function. To understand cell-material interaction, we determined the porosity, biocompatibility, and stiffness of the ECM mimics. The ECM mimics have median pore sizes of 10.7 and 13.3 μm, close to the average nucleus size of CTLs (~8.6 μm), and good biocompatibility to facilitate cell migration. The stiffness of the ECM mimics corresponds to biologically relevant microenvironments such as lungs and kidneys. Using time-lapse fluorescence microscopy, 3D cell migration was imaged and measured. CTLs migrated faster in softer ECM mimic with larger pores, consistent with previous studies in collagen (the gold standard ECM mimic for CTLs). The work herein demonstrates that the PEG-based ECM mimic can serve as an in vitro tool to elucidate the cell dynamics of CTLs. Thus, this study opens possibilities to study the mechanics of CTLs in well-defined ECM mimic conditions in vitro.
Collapse
Affiliation(s)
- M A Kristine Tolentino
- School of Chemistry and Australian Centre of NanoMedicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Mir Hadi Seyedzadeh
- European Molecular Biology Laboratory Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Newton Gil Peres
- European Molecular Biology Laboratory Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Eric Yiwei Du
- School of Chemistry and Australian Centre of NanoMedicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Lin Zhu
- School of Chemistry and Australian Centre of NanoMedicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Katharina Gaus
- European Molecular Biology Laboratory Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jesse Goyette
- European Molecular Biology Laboratory Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - J Justin Gooding
- School of Chemistry and Australian Centre of NanoMedicine, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
4
|
Marzban S, Srivastava S, Kartika S, Bravo R, Safriel R, Zarski A, Anderson ARA, Chung CH, Amelio AL, West J. Spatial interactions modulate tumor growth and immune infiltration. NPJ Syst Biol Appl 2024; 10:106. [PMID: 39349537 PMCID: PMC11442770 DOI: 10.1038/s41540-024-00438-1] [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: 05/10/2024] [Accepted: 09/10/2024] [Indexed: 10/02/2024] Open
Abstract
Direct observation of tumor-immune interactions is unlikely in tumors with currently available technology, but computational simulations based on clinical data can provide insight to test hypotheses. It is hypothesized that patterns of collagen evolve as a mechanism of immune escape, but the exact nature of immune-collagen interactions is poorly understood. Spatial data quantifying collagen fiber alignment in squamous cell carcinomas indicates that late-stage disease is associated with highly aligned fibers. Our computational modeling framework discriminates between two hypotheses: immune cell migration that moves (1) parallel or (2) perpendicular to collagen fiber orientation. The modeling recapitulates immune-extracellular matrix interactions where collagen patterns provide immune protection, leading to an emergent inverse relationship between disease stage and immune coverage. Here, computational modeling provides important mechanistic insights by defining a kernel cell-cell interaction function that considers a spectrum of local (cell-scale) to global (tumor-scale) spatial interactions. Short-range interaction kernels provide a mechanism for tumor cell survival under conditions with strong Allee effects, while asymmetric tumor-immune interaction kernels lead to poor immune response. Thus, the length scale of tumor-immune interaction kernels drives tumor growth and infiltration.
Collapse
Affiliation(s)
- Sadegh Marzban
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Sonal Srivastava
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Sharon Kartika
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Kolkata, India
| | - Rafael Bravo
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Rachel Safriel
- High School Internship Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Aidan Zarski
- High School Internship Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Alexander R A Anderson
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Christine H Chung
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Antonio L Amelio
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jeffrey West
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA.
| |
Collapse
|
5
|
Cañellas-Socias A, Sancho E, Batlle E. Mechanisms of metastatic colorectal cancer. Nat Rev Gastroenterol Hepatol 2024; 21:609-625. [PMID: 38806657 DOI: 10.1038/s41575-024-00934-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2024] [Indexed: 05/30/2024]
Abstract
Despite extensive research and improvements in understanding colorectal cancer (CRC), its metastatic form continues to pose a substantial challenge, primarily owing to limited therapeutic options and a poor prognosis. This Review addresses the emerging focus on metastatic CRC (mCRC), which has historically been under-studied compared with primary CRC despite its lethality. We delve into two crucial aspects: the molecular and cellular determinants facilitating CRC metastasis and the principles guiding the evolution of metastatic disease. Initially, we examine the genetic alterations integral to CRC metastasis, connecting them to clinically marked characteristics of advanced CRC. Subsequently, we scrutinize the role of cellular heterogeneity and plasticity in metastatic spread and therapy resistance. Finally, we explore how the tumour microenvironment influences metastatic disease, emphasizing the effect of stromal gene programmes and the immune context. The ongoing research in these fields holds immense importance, as its future implications are projected to revolutionize the treatment of patients with mCRC, hopefully offering a promising outlook for their survival.
Collapse
Affiliation(s)
- Adrià Cañellas-Socias
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain.
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| | - Elena Sancho
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
6
|
Inamdar VV, Hao S, Stephan SB, Stephan MT. Biomaterial-based scaffolds for direct in situ programming of tumor-infiltrating T lymphocytes. J Control Release 2024; 370:310-317. [PMID: 38677524 DOI: 10.1016/j.jconrel.2024.04.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/14/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
Adoptive cell therapy with tumor-infiltrating T cells (TILs) has generated exciting clinical trial results for the treatment of unresectable solid tumors. However, solid tumors remain difficult targets for adoptively transferred T cells, due in part to poor migration of TILs to the tumor, physical barriers to infiltration, and active suppression of TILs by the tumor. Furthermore, a highly skilled team is required to obtain tumor tissue, isolate and expand the TILs ex vivo, and reinfuse them into the patient, which drives up costs and limits patient access. Here, we describe a cell-free polymer implant designed to recruit, genetically reprogram and expand host T cells at tumor lesions in situ. Importantly, the scaffold can be fabricated on a large scale and is stable to lyophilization. Using a mouse breast cancer model, we show that the implants quickly and efficiently amass cancer-specific host lymphocytes at the tumor site in quantities sufficient to bring about long-term tumor regression. Given that surgical care is the mainstay of cancer treatment for many patients, this technology could be easily implemented in a clinical setting as an add-on to surgery for solid tumors. Furthermore, the approach could be broadened to recruit and genetically reprogram other therapeutically desirable host cells, such as macrophages, natural killer cells or dendritic cells, potentially boosting the antitumor effectiveness of the implant even more.
Collapse
Affiliation(s)
- V V Inamdar
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington 98109, USA
| | - S Hao
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington 98109, USA
| | - S B Stephan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington 98109, USA
| | - M T Stephan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington 98109, USA; Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, Washington 98195, USA; Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington 98195, USA.
| |
Collapse
|
7
|
Chen B, Liu J. Prospects and challenges of CAR-T in the treatment of ovarian cancer. Int Immunopharmacol 2024; 133:112112. [PMID: 38640714 DOI: 10.1016/j.intimp.2024.112112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Ovarian cancer ranks as the seventh most prevalent cancer among women and is considered the most lethal gynecological malignancy on a global scale. The absence of reliable screening techniques, coupled with the insidious onset of nonspecific symptoms, often results in a delayed diagnosis, typically at an advanced stage characterized by peritoneal involvement. Management of advanced tumors typically involves a combination of chemotherapy and cytoreductive surgery. However, the therapeutic arsenal for ovarian cancer patients remains limited, highlighting the unmet need for precise, targeted, and sustained-release pharmacological agents. Genetically engineered T cells expressing chimeric antigen receptors (CARs) represent a promising novel therapeutic modality that selectively targets specific antigens, demonstrating robust and enduring antitumor responses in numerous patients. CAR T cell therapy has exhibited notable efficacy in hematological malignancies and is currently under investigation for its potential in treating various solid tumors, including ovarian cancer. Currently, numerous researchers are engaged in the development of novel CAR-T cells designed to target ovarian cancer, with subsequent evaluation of these candidate cells in preclinical studies. Given the ability of chimeric antigen receptor (CAR) expressing T cells to elicit potent and long-lasting anti-tumor effects, this therapeutic approach holds significant promise for the treatment of ovarian cancer. This review article examines the utilization of CAR-T cells in the context of ovarian cancer therapy.
Collapse
Affiliation(s)
- Biqing Chen
- Harbin Medical University, Harbin, Heilongjiang, China.
| | | |
Collapse
|
8
|
Marzban S, Srivastava S, Kartika S, Bravo R, Safriel R, Zarski A, Anderson A, Chung CH, Amelio AL, West J. Spatial interactions modulate tumor growth and immune infiltration. RESEARCH SQUARE 2024:rs.3.rs-3962451. [PMID: 38826398 PMCID: PMC11142313 DOI: 10.21203/rs.3.rs-3962451/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Lenia, a cellular automata framework used in artificial life, provides a natural setting to implement mathematical models of cancer incorporating features such as morphogenesis, homeostasis, motility, reproduction, growth, stimuli response, evolvability, and adaptation. Historically, agent-based models of cancer progression have been constructed with rules that govern birth, death and migration, with attempts to map local rules to emergent global growth dynamics. In contrast, Lenia provides a flexible framework for considering a spectrum of local (cell-scale) to global (tumor-scale) dynamics by defining an interaction kernel governing density-dependent growth dynamics. Lenia can recapitulate a range of cancer model classifications including local or global, deterministic or stochastic, non-spatial or spatial, single or multi-population, and off or on-lattice. Lenia is subsequently used to develop data-informed models of 1) single-population growth dynamics, 2) multi-population cell-cell competition models, and 3) cell migration or chemotaxis. Mathematical modeling provides important mechanistic insights. First, short-range interaction kernels provide a mechanism for tumor cell survival under conditions with strong Allee effects. Next, we find that asymmetric interaction tumor-immune kernels lead to poor immune response. Finally, modeling recapitulates immune-ECM interactions where patterns of collagen formation provide immune protection, indicated by an emergent inverse relationship between disease stage and immune coverage.
Collapse
Affiliation(s)
- Sadegh Marzban
- Integrated Mathematical Oncology Dept., H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Sonal Srivastava
- Dept. of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Sharon Kartika
- Dept. of Biological Sciences, Indian Institute of Science Education and Research Kolkata
| | - Rafael Bravo
- Integrated Mathematical Oncology Dept., H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Rachel Safriel
- High School Internship Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Aidan Zarski
- High School Internship Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Alexander Anderson
- Integrated Mathematical Oncology Dept., H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Christine H. Chung
- Dept. of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Antonio L. Amelio
- Dept. of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Dept. of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Jeffrey West
- Integrated Mathematical Oncology Dept., H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| |
Collapse
|
9
|
Skirzynska A, Xue C, Shoichet MS. Engineering Biomaterials to Model Immune-Tumor Interactions In Vitro. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310637. [PMID: 38349174 DOI: 10.1002/adma.202310637] [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: 10/12/2023] [Revised: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Engineered biomaterial scaffolds are becoming more prominent in research laboratories to study drug efficacy for oncological applications in vitro, but do they have a place in pharmaceutical drug screening pipelines? The low efficacy of cancer drugs in phase II/III clinical trials suggests that there are critical mechanisms not properly accounted for in the pre-clinical evaluation of drug candidates. Immune cells associated with the tumor may account for some of these failures given recent successes with cancer immunotherapies; however, there are few representative platforms to study immune cells in the context of cancer as traditional 2D culture is typically monocultures and humanized animal models have a weakened immune composition. Biomaterials that replicate tumor microenvironmental cues may provide a more relevant model with greater in vitro complexity. In this review, the authors explore the pertinent microenvironmental cues that drive tumor progression in the context of the immune system, discuss how these cues can be incorporated into hydrogel design to culture immune cells, and describe progress toward precision oncological drug screening with engineered tissues.
Collapse
Affiliation(s)
- Arianna Skirzynska
- Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
| | - Chang Xue
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
- Institute for Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
| | - Molly S Shoichet
- Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
- Institute for Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Department of Chemistry, University of Toronto, 80 College Street, Toronto, ON, M5S 3H4, Canada
| |
Collapse
|
10
|
Borst R, Meyaard L, Pascoal Ramos MI. Understanding the matrix: collagen modifications in tumors and their implications for immunotherapy. J Transl Med 2024; 22:382. [PMID: 38659022 PMCID: PMC11040975 DOI: 10.1186/s12967-024-05199-3] [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: 12/15/2023] [Accepted: 04/13/2024] [Indexed: 04/26/2024] Open
Abstract
Tumors are highly complex and heterogenous ecosystems where malignant cells interact with healthy cells and the surrounding extracellular matrix (ECM). Solid tumors contain large ECM deposits that can constitute up to 60% of the tumor mass. This supports the survival and growth of cancerous cells and plays a critical role in the response to immune therapy. There is untapped potential in targeting the ECM and cell-ECM interactions to improve existing immune therapy and explore novel therapeutic strategies. The most abundant proteins in the ECM are the collagen family. There are 28 different collagen subtypes that can undergo several post-translational modifications (PTMs), which alter both their structure and functionality. Here, we review current knowledge on tumor collagen composition and the consequences of collagen PTMs affecting receptor binding, cell migration and tumor stiffness. Furthermore, we discuss how these alterations impact tumor immune responses and how collagen could be targeted to treat cancer.
Collapse
Affiliation(s)
- Rowie Borst
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - M Ines Pascoal Ramos
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
11
|
Lahusen A, Cai J, Schirmbeck R, Wellstein A, Kleger A, Seufferlein T, Eiseler T, Lin YN. A pancreatic cancer organoid-in-matrix platform shows distinct sensitivities to T cell killing. Sci Rep 2024; 14:9377. [PMID: 38654067 DOI: 10.1038/s41598-024-60107-5] [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: 09/17/2023] [Accepted: 04/18/2024] [Indexed: 04/25/2024] Open
Abstract
Poor treatment responses of pancreatic ductal adenocarcinoma (PDAC) are in large part due to tumor heterogeneity and an immunosuppressive desmoplastic tumor stroma that impacts interactions with cells in the tumor microenvironment (TME). Thus, there is a pressing need for models to probe the contributions of cellular and noncellular crosstalk. Organoids are promising model systems with the potential to generate a plethora of data including phenotypic, transcriptomic and genomic characterization but still require improvements in culture conditions mimicking the TME. Here, we describe an INTERaction with Organoid-in-MatriX ("InterOMaX") model system, that presents a 3D co-culture-based platform for investigating matrix-dependent cellular crosstalk. We describe its potential to uncover new molecular mechanisms of T cell responses to murine KPC (LSL-KrasG12D/+27/Trp53tm1Tyj/J/p48Cre/+) PDAC cells as well as PDAC patient-derived organoids (PDOs). For this, a customizable matrix and homogenously sized organoid-in-matrix positioning of cancer cells were designed based on a standardized agarose microwell chip array system and established for co-culture with T cells and inclusion of stromal cells. We describe the detection and orthogonal analysis of murine and human PDAC cell populations with distinct sensitivity to T cell killing that is corroborated in vivo. By enabling both identification and validation of gene candidates for T cell resistance, this platform sets the stage for better mechanistic understanding of cancer cell-intrinsic resistance phenotypes in PDAC.
Collapse
Affiliation(s)
- Anton Lahusen
- Department of Internal Medicine I, Gastroenterology, Endocrinology, Nephrology, Nutrition and Metabolism, Ulm University Hospital, Albert Einstein Allee 23, 89081, Ulm, Germany
| | - Jierui Cai
- Department of Internal Medicine I, Gastroenterology, Endocrinology, Nephrology, Nutrition and Metabolism, Ulm University Hospital, Albert Einstein Allee 23, 89081, Ulm, Germany
| | - Reinhold Schirmbeck
- Department of Internal Medicine I, Gastroenterology, Endocrinology, Nephrology, Nutrition and Metabolism, Ulm University Hospital, Albert Einstein Allee 23, 89081, Ulm, Germany
| | - Anton Wellstein
- Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road NW, Washington, DC, 20007, USA
| | - Alexander Kleger
- Department of Internal Medicine I, Gastroenterology, Endocrinology, Nephrology, Nutrition and Metabolism, Ulm University Hospital, Albert Einstein Allee 23, 89081, Ulm, Germany
- Institute of Molecular Oncology and Stem Cell Biology (IMOS), Ulm University Hospital, 89081, Ulm, Germany
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine I, Ulm University Hospital, 89081, Ulm, Germany
- Organoid Core Facility, Ulm University Hospital, 89081, Ulm, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine I, Gastroenterology, Endocrinology, Nephrology, Nutrition and Metabolism, Ulm University Hospital, Albert Einstein Allee 23, 89081, Ulm, Germany
| | - Tim Eiseler
- Department of Internal Medicine I, Gastroenterology, Endocrinology, Nephrology, Nutrition and Metabolism, Ulm University Hospital, Albert Einstein Allee 23, 89081, Ulm, Germany
| | - Yuan-Na Lin
- Department of Internal Medicine I, Gastroenterology, Endocrinology, Nephrology, Nutrition and Metabolism, Ulm University Hospital, Albert Einstein Allee 23, 89081, Ulm, Germany.
| |
Collapse
|
12
|
Yapp C, Nirmal AJ, Zhou F, Maliga Z, Tefft JB, Llopis PM, Murphy GF, Lian CG, Danuser G, Santagata S, Sorger PK. Multiplexed 3D Analysis of Immune States and Niches in Human Tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.10.566670. [PMID: 38014052 PMCID: PMC10680601 DOI: 10.1101/2023.11.10.566670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Tissue homeostasis and the emergence of disease are controlled by changes in the proportions of resident and recruited cells, their organization into cellular neighbourhoods, and their interactions with acellular tissue components. Highly multiplexed tissue profiling (spatial omics)1 makes it possible to study this microenvironment in situ, usually in 4-5 micron thick sections (the standard histopathology format)2. Microscopy-based tissue profiling is commonly performed at a resolution sufficient to determine cell types but not to detect subtle morphological features associated with cytoskeletal reorganisation, juxtracrine signalling, or membrane trafficking3. Here we describe a high-resolution 3D imaging approach able to characterize a wide variety of organelles and structures at sub-micron scale while simultaneously quantifying millimetre-scale spatial features. This approach combines cyclic immunofluorescence (CyCIF) imaging4 of over 50 markers with confocal microscopy of archival human tissue thick enough (30-40 microns) to fully encompass two or more layers of intact cells. 3D imaging of entire cell volumes substantially improves the accuracy of cell phenotyping and allows cell proximity to be scored using plasma membrane apposition, not just nuclear position. In pre-invasive melanoma in situ5, precise phenotyping shows that adjacent melanocytic cells are plastic in state and participate in tightly localised niches of interferon signalling near sites of initial invasion into the underlying dermis. In this and metastatic melanoma, mature and precursor T cells engage in an unexpectedly diverse array of juxtracrine and membrane-membrane interactions as well as looser "neighbourhood" associations6 whose morphologies reveal functional states. These data provide new insight into the transitions occurring during early tumour formation and immunoediting and demonstrate the potential for phenotyping of tissues at a level of detail previously restricted to cultured cells and organoids.
Collapse
Affiliation(s)
- Clarence Yapp
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
- Ludwig Centre at Harvard, Harvard Medical School, Boston, MA, 02115, USA
| | - Ajit J. Nirmal
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
- Ludwig Centre at Harvard, Harvard Medical School, Boston, MA, 02115, USA
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Felix Zhou
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Zoltan Maliga
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Juliann B. Tefft
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
- Ludwig Centre at Harvard, Harvard Medical School, Boston, MA, 02115, USA
| | - Paula Montero Llopis
- Microscopy Resources on the North Quad (MicRoN), Harvard Medical School, Boston, MA 02115, USA
| | - George F. Murphy
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Christine G. Lian
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sandro Santagata
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
- Ludwig Centre at Harvard, Harvard Medical School, Boston, MA, 02115, USA
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | - Peter K. Sorger
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
- Ludwig Centre at Harvard, Harvard Medical School, Boston, MA, 02115, USA
- Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | | |
Collapse
|
13
|
Johnston AC, Alicea GM, Lee CC, Patel PV, Hanna EA, Vaz E, Forjaz A, Wan Z, Nair PR, Lim Y, Chen T, Du W, Kim D, Nichakawade TD, Rebecca VW, Bonifant CL, Fan R, Kiemen AL, Wu PH, Wirtz D. Engineering self-propelled tumor-infiltrating CAR T cells using synthetic velocity receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.13.571595. [PMID: 38168186 PMCID: PMC10760159 DOI: 10.1101/2023.12.13.571595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Chimeric antigen receptor (CAR) T cells express antigen-specific synthetic receptors, which upon binding to cancer cells, elicit T cell anti-tumor responses. CAR T cell therapy has enjoyed success in the clinic for hematological cancer indications, giving rise to decade-long remissions in some cases. However, CAR T therapy for patients with solid tumors has not seen similar success. Solid tumors constitute 90% of adult human cancers, representing an enormous unmet clinical need. Current approaches do not solve the central problem of limited ability of therapeutic cells to migrate through the stromal matrix. We discover that T cells at low and high density display low- and high-migration phenotypes, respectively. The highly migratory phenotype is mediated by a paracrine pathway from a group of self-produced cytokines that include IL5, TNFα, IFNγ, and IL8. We exploit this finding to "lock-in" a highly migratory phenotype by developing and expressing receptors, which we call velocity receptors (VRs). VRs target these cytokines and signal through these cytokines' cognate receptors to increase T cell motility and infiltrate lung, ovarian, and pancreatic tumors in large numbers and at doses for which control CAR T cells remain confined to the tumor periphery. In contrast to CAR therapy alone, VR-CAR T cells significantly attenuate tumor growth and extend overall survival. This work suggests that approaches to the design of immune cell receptors that focus on migration signaling will help current and future CAR cellular therapies to infiltrate deep into solid tumors.
Collapse
Affiliation(s)
- Adrian C Johnston
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | | | - Cameron C Lee
- Department of Biomedical Engineering, Johns Hopkins University
| | - Payal V Patel
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - Eban A Hanna
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - Eduarda Vaz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - André Forjaz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - Zeqi Wan
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University
| | - Praful R Nair
- Institute for NanoBioTechnology, Johns Hopkins University
| | - Yeongseo Lim
- Department of Biomedical Engineering, Johns Hopkins University
| | - Tina Chen
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - Wenxuan Du
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - Dongjoo Kim
- Department of Biomedical Engineering, Yale University
| | - Tushar D Nichakawade
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University
| | - Vito W Rebecca
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University
| | - Challice L Bonifant
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University
| | - Rong Fan
- Department of Biomedical Engineering, Yale University
| | - Ashley L Kiemen
- Institute for NanoBioTechnology, Johns Hopkins University
- Department of Pathology, Johns Hopkins School of Medicine, Johns Hopkins University
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University
| | - Pei-Hsun Wu
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
| | - Denis Wirtz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
- Department of Pathology, Johns Hopkins School of Medicine, Johns Hopkins University
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University
| |
Collapse
|
14
|
Marzban S, Srivastava S, Kartika S, Bravo R, Safriel R, Zarski A, Anderson A, Chung CH, Amelio AL, West J. Spatial interactions modulate tumor growth and immune infiltration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575036. [PMID: 38370722 PMCID: PMC10871273 DOI: 10.1101/2024.01.10.575036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Direct observation of immune cell trafficking patterns and tumor-immune interactions is unlikely in human tumors with currently available technology, but computational simulations based on clinical data can provide insight to test hypotheses. It is hypothesized that patterns of collagen formation evolve as a mechanism of immune escape, but the exact nature of the interaction between immune cells and collagen is poorly understood. Spatial data quantifying the degree of collagen fiber alignment in squamous cell carcinomas indicates that late stage disease is associated with highly aligned fibers. Here, we introduce a computational modeling framework (called Lenia) to discriminate between two hypotheses: immune cell migration that moves 1) parallel or 2) perpendicular to collagen fiber orientation. The modeling recapitulates immune-ECM interactions where collagen patterns provide immune protection, leading to an emergent inverse relationship between disease stage and immune coverage. We also illustrate the capabilities of Lenia to model the evolution of tumor progression and immune predation. Lenia provides a flexible framework for considering a spectrum of local (cell-scale) to global (tumor-scale) dynamics by defining a kernel cell-cell interaction function that governs tumor growth dynamics under immune predation with immune cell migration. Mathematical modeling provides important mechanistic insights into cell interactions. Short-range interaction kernels provide a mechanism for tumor cell survival under conditions with strong Allee effects, while asymmetric tumor-immune interaction kernels lead to poor immune response. Thus, the length scale of tumor-immune interactions drives tumor growth and infiltration.
Collapse
Affiliation(s)
- Sadegh Marzban
- Integrated Mathematical Oncology Dept., H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Sonal Srivastava
- Dept. of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Sharon Kartika
- Dept. of Biological Sciences, Indian Institute of Science Education and Research Kolkata
| | - Rafael Bravo
- Integrated Mathematical Oncology Dept., H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Rachel Safriel
- High School Internship Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Aidan Zarski
- High School Internship Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Alexander Anderson
- Integrated Mathematical Oncology Dept., H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Christine H. Chung
- Dept. of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Antonio L. Amelio
- Dept. of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Dept. of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Jeffrey West
- Integrated Mathematical Oncology Dept., H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| |
Collapse
|
15
|
Jeffreys N, Brockman JM, Zhai Y, Ingber DE, Mooney DJ. Mechanical forces amplify TCR mechanotransduction in T cell activation and function. APPLIED PHYSICS REVIEWS 2024; 11:011304. [PMID: 38434676 PMCID: PMC10848667 DOI: 10.1063/5.0166848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/08/2023] [Indexed: 03/05/2024]
Abstract
Adoptive T cell immunotherapies, including engineered T cell receptor (eTCR) and chimeric antigen receptor (CAR) T cell immunotherapies, have shown efficacy in treating a subset of hematologic malignancies, exhibit promise in solid tumors, and have many other potential applications, such as in fibrosis, autoimmunity, and regenerative medicine. While immunoengineering has focused on designing biomaterials to present biochemical cues to manipulate T cells ex vivo and in vivo, mechanical cues that regulate their biology have been largely underappreciated. This review highlights the contributions of mechanical force to several receptor-ligand interactions critical to T cell function, with central focus on the TCR-peptide-loaded major histocompatibility complex (pMHC). We then emphasize the role of mechanical forces in (i) allosteric strengthening of the TCR-pMHC interaction in amplifying ligand discrimination during T cell antigen recognition prior to activation and (ii) T cell interactions with the extracellular matrix. We then describe approaches to design eTCRs, CARs, and biomaterials to exploit TCR mechanosensitivity in order to potentiate T cell manufacturing and function in adoptive T cell immunotherapy.
Collapse
Affiliation(s)
| | | | - Yunhao Zhai
- Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
16
|
Pally D, Naba A. Extracellular matrix dynamics: A key regulator of cell migration across length-scales and systems. Curr Opin Cell Biol 2024; 86:102309. [PMID: 38183892 PMCID: PMC10922734 DOI: 10.1016/j.ceb.2023.102309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/11/2023] [Accepted: 12/11/2023] [Indexed: 01/08/2024]
Abstract
The interactions between cells and their surrounding extracellular matrix (ECM) are dynamic and play critical roles in cell migration during development, health, and diseases. Recent advances have highlighted the complexity and diversity of ECM compositions, or "matrisomes", of tissues resulting in ECMs of different physical, mechanical, and biochemical properties. Investigating the effects of these properties on cell-ECM interactions in the context of cell migration have led to a better understanding of the principles underlying tissue morphogenesis, wound healing, immune response, or cancer metastasis. These new insights into the interplay between ECM dynamics and cell migration can lead to the identification of unique opportunities for therapeutic interventions.
Collapse
Affiliation(s)
- Dharma Pally
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA; University of Illinois Cancer Center, Chicago, IL 60612, USA.
| |
Collapse
|
17
|
Ozulumba T, Zatorski JM, Arneja A, Hammel JH, Braciale TJ, Luckey CJ, Munson JM, Pompano RR. Mitigating reactive oxygen species production and increasing gel porosity improves lymphocyte motility and fibroblast spreading in photocrosslinked gelatin-thiol hydrogels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.14.574282. [PMID: 38293038 PMCID: PMC10827049 DOI: 10.1101/2024.01.14.574282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
On-chip 3D culture systems that incorporate immune cells such as lymphocytes and stromal cells are needed to model immune organs in engineered systems such as organs-on-chip. Photocrosslinking is a useful tool for creating such immune-competent hydrogel cultures with spatial cell organization. However, loss of viability and motility in photocrosslinked gels can limit its utility, especially when working with fragile primary cells. We hypothesized that optimizing photoexposure-induced ROS production, hydrogel porosity or a combination of both factors was necessary to sustain cell viability and motility during culture in photocrosslinked gelatin-thiol (GelSH) hydrogels. Jurkat T cells, primary human CD4+ T cells and human lymphatic fibroblasts were selected as representative lymphoid immune cells to test this hypothesis. Direct exposure of these cells to 385 nm light and LAP photoinitiator dramatically increased ROS levels. Pretreatment with an antioxidant, ascorbic acid (AA), protected the cells from light + LAP-induced ROS and was non-toxic at optimized doses. Furthermore, scanning electron microscopy showed that native GelSH hydrogels had limited porosity, and that adding collagen to GelSH precursor before crosslinking markedly increased gel porosity. Next, we tested the impact of AA pretreatment and increasing gel porosity, alone or in combination, on cell viability and function in 3D GelSH hydrogel cultures. Increasing gel porosity, rather than AA pretreatment, was more critical for rescuing viability of Jurkat T cells and spreading of human lymphatic fibroblasts in GelSH-based gels, but both factors improved the motility of primary human CD4+ T cells. Increased porosity enabled formation of spatially organized co-cultures of primary human CD4+ T cells and human lymphatic fibroblasts in photo-crosslinked gels in a multi-lane microfluidic chip, towards modeling the lymphoid organ microenvironment. Some optimization is still needed to improve homogeneity between regions on the chip. These findings will enable researchers utilizing photocrosslinking methods to develop immunocompetent 3D culture models that support viability and function of sensitive lymphoid cells.
Collapse
|
18
|
Zhovmer AS, Manning A, Smith C, Nguyen A, Prince O, Sáez PJ, Ma X, Tsygankov D, Cartagena-Rivera AX, Singh NA, Singh RK, Tabdanov ED. Septins provide microenvironment sensing and cortical actomyosin partitioning in motile amoeboid T lymphocytes. SCIENCE ADVANCES 2024; 10:eadi1788. [PMID: 38170778 DOI: 10.1126/sciadv.adi1788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
The all-terrain motility of lymphocytes in tissues and tissue-like gels is best described as amoeboid motility. For amoeboid motility, lymphocytes do not require specific biochemical or structural modifications to the surrounding extracellular matrix. Instead, they rely on changing shape and steric interactions with the microenvironment. However, the exact mechanism of amoeboid motility remains elusive. Here, we report that septins participate in amoeboid motility of T cells, enabling the formation of F-actin and α-actinin-rich cortical rings at the sites of cell cortex-indenting collisions with the extracellular matrix. Cortical rings compartmentalize cells into chains of spherical segments that are spatially conformed to the available lumens, forming transient "hourglass"-shaped steric locks onto the surrounding collagen fibers. The steric lock facilitates pressure-driven peristaltic propulsion of cytosolic content by individually contracting cell segments. Our results suggest that septins provide microenvironment-guided partitioning of actomyosin contractility and steric pivots required for amoeboid motility of T cells in tissue-like microenvironments.
Collapse
Affiliation(s)
- Alexander S Zhovmer
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Alexis Manning
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Chynna Smith
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Ashley Nguyen
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Olivia Prince
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Pablo J Sáez
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, and Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Xuefei Ma
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Denis Tsygankov
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Alexander X Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Niloy A Singh
- Department of Hematology Oncology, University of Rochester Medical Center, Rochester, NY, USA
| | - Rakesh K Singh
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Erdem D Tabdanov
- Department of Pharmacology, Penn State College of Medicine, The Pennsylvania State University, Hershey-Hummelstown, PA, USA
- Penn State Cancer Institute, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| |
Collapse
|
19
|
Pethő Z, Najder K, Beel S, Fels B, Neumann I, Schimmelpfennig S, Sargin S, Wolters M, Grantins K, Wardelmann E, Mitkovski M, Oeckinghaus A, Schwab A. Acid-base homeostasis orchestrated by NHE1 defines the pancreatic stellate cell phenotype in pancreatic cancer. JCI Insight 2023; 8:e170928. [PMID: 37643024 PMCID: PMC10619433 DOI: 10.1172/jci.insight.170928] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) progresses in an organ with a unique pH landscape, where the stroma acidifies after each meal. We hypothesized that disrupting this pH landscape during PDAC progression triggers pancreatic stellate cells (PSCs) and cancer-associated fibroblasts (CAFs) to induce PDAC fibrosis. We revealed that alkaline environmental pH was sufficient to induce PSC differentiation to a myofibroblastic phenotype. We then mechanistically dissected this finding, focusing on the involvement of the Na+/H+ exchanger NHE1. Perturbing cellular pH homeostasis by inhibiting NHE1 with cariporide partially altered the myofibroblastic PSC phenotype. To show the relevance of this finding in vivo, we targeted NHE1 in murine PDAC (KPfC). Indeed, tumor fibrosis decreased when mice received the NHE1-inhibitor cariporide in addition to gemcitabine treatment. Moreover, the tumor immune infiltrate shifted from granulocyte rich to more lymphocytic. Taken together, our study provides mechanistic evidence on how the pancreatic pH landscape shapes pancreatic cancer through tuning PSC differentiation.
Collapse
Affiliation(s)
| | | | - Stephanie Beel
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| | - Benedikt Fels
- Institute of Physiology II and
- Institute of Physiology, University of Lübeck, Lübeck, Germany
| | | | | | | | - Maria Wolters
- Gerhard-Domagk-Institute of Pathology, University of Münster, Münster, Germany
| | - Klavs Grantins
- Gerhard-Domagk-Institute of Pathology, University of Münster, Münster, Germany
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, University of Münster, Münster, Germany
| | - Miso Mitkovski
- City Campus Light Microscopy Facility, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
| | - Andrea Oeckinghaus
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| | | |
Collapse
|
20
|
Shi W, Mirza S, Kuss M, Liu B, Hartin A, Wan S, Kong Y, Mohapatra B, Krishnan M, Band H, Band V, Duan B. Embedded Bioprinting of Breast Tumor Cells and Organoids Using Low-Concentration Collagen-Based Bioinks. Adv Healthc Mater 2023; 12:e2300905. [PMID: 37422447 PMCID: PMC10592394 DOI: 10.1002/adhm.202300905] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/10/2023]
Abstract
Bioinks for 3D bioprinting of tumor models should not only meet printability requirements but also accurately maintain and support phenotypes of tumor surrounding cells to recapitulate key tumor hallmarks. Collagen is a major extracellular matrix protein for solid tumors, but low viscosity of collagen solution has made 3D bioprinted cancer models challenging. This work produces embedded, bioprinted breast cancer cells and tumor organoid models using low-concentration collagen I based bioinks. The biocompatible and physically crosslinked silk fibroin hydrogel is used to generate the support bath for the embedded 3D printing. The composition of the collagen I based bioink is optimized with a thermoresponsive hyaluronic acid-based polymer to maintain the phenotypes of both the noninvasive epithelial and invasive breast cancer cells, as well as cancer-associated fibroblasts. Mouse breast tumor organoids are bioprinted using optimized collagen bioink to mimic in vivo tumor morphology. A vascularized tumor model is also created using a similar strategy, with significantly enhanced vasculature formation under hypoxia. This study shows the great potential of embedded bioprinted breast tumor models utilizing a low-concentration collagen-based bioink for advancing the understanding of tumor cell biology and facilitating drug discovery research.
Collapse
Affiliation(s)
- Wen Shi
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Sameer Mirza
- Department of GeneticsCell Biology and AnatomyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of ChemistryCollege of ScienceUnited Arab Emirates UniversityAbu DhabiUnited Arab Emirates
| | - Mitchell Kuss
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bo Liu
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Andrew Hartin
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Shibiao Wan
- Department of GeneticsCell Biology and AnatomyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Yunfan Kong
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bhopal Mohapatra
- Department of GeneticsCell Biology and AnatomyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Mena Krishnan
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Hamid Band
- Eppley InstituteUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Vimla Band
- Department of GeneticsCell Biology and AnatomyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bin Duan
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of SurgeryUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of Mechanical EngineeringUniversity of Nebraska–LincolnLincolnNE68588USA
| |
Collapse
|
21
|
Clémençon P, Alpern S, Gal S, Casas J. Counterintuitive prey strategies against predators with finite budgets: protection heterogeneity among sites matters more than their number. J R Soc Interface 2023; 20:20230216. [PMID: 37727074 PMCID: PMC10509579 DOI: 10.1098/rsif.2023.0216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023] Open
Abstract
Combining the search and pursuit aspects of predator-prey interactions into a single game, where the payoff to the Searcher (predator) is the probability of finding and capturing the Hider (prey) within a fixed number of searches was proposed by Gal and Casas (J. R. Soc. Interface 11, 20140062 (doi:10.1098/rsif.2014.0062)). Subsequent models allowed the predator to continue its search (in another 'round') if the prey was found but escaped the chase. However, it is unrealistic to allow this pattern of prey relocation to go on forever, so here we introduce a limit of the total number of searches, in all 'rounds', that the predator can carry out. We show how habitat structural complexity affects the mean time until capture: the quality of the location with the lowest capture probability matters more than the number of hiding locations. Moreover, we observed that the parameter space defined by the capture probabilities in each location and the budget of the predator can be divided into distinct domains, defining whether the prey ought to play with pure or mixed hiding strategies.
Collapse
Affiliation(s)
- Paul Clémençon
- Insect Biology Research Institute, UMR 7261 CNRS / University of Tours, Tours, France
| | - Steve Alpern
- Warwick Business School, University of Warwick, Coventry, UK
| | - Shmuel Gal
- Department of Statistics, University of Haifa, Haifa, Israel
| | - Jérôme Casas
- Insect Biology Research Institute, UMR 7261 CNRS / University of Tours, Tours, France
| |
Collapse
|
22
|
Chia DKA, Demuytere J, Ernst S, Salavati H, Ceelen W. Effects of Hyperthermia and Hyperthermic Intraperitoneal Chemoperfusion on the Peritoneal and Tumor Immune Contexture. Cancers (Basel) 2023; 15:4314. [PMID: 37686590 PMCID: PMC10486595 DOI: 10.3390/cancers15174314] [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: 07/22/2023] [Revised: 08/12/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Hyperthermia combined with intraperitoneal (IP) drug delivery is increasingly used in the treatment of peritoneal metastases (PM). Hyperthermia enhances tumor perfusion and increases drug penetration after IP delivery. The peritoneum is increasingly recognized as an immune-privileged organ with its own distinct immune microenvironment. Here, we review the immune landscape of the healthy peritoneal cavity and immune contexture of peritoneal metastases. Next, we review the potential benefits and unwanted tumor-promoting effects of hyperthermia and the associated heat shock response on the tumor immune microenvironment. We highlight the potential modulating effect of hyperthermia on the biomechanical properties of tumor tissue and the consequences for immune cell infiltration. Data from translational and clinical studies are reviewed. We conclude that (mild) hyperthermia and HIPEC have the potential to enhance antitumor immunity, but detailed further studies are required to distinguish beneficial from tumor-promoting effects.
Collapse
Affiliation(s)
- Daryl K. A. Chia
- Department of Surgery, National University Hospital, National University Health System, Singapore 119074, Singapore
| | - Jesse Demuytere
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Sam Ernst
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Hooman Salavati
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Wim Ceelen
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Department of GI Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
23
|
Senosain MF, Zou Y, Patel K, Zhao S, Coullomb A, Rowe DJ, Lehman JM, Irish JM, Maldonado F, Kammer MN, Pancaldi V, Lopez CF. Integrated Multi-omics Analysis of Early Lung Adenocarcinoma Links Tumor Biological Features with Predicted Indolence or Aggressiveness. CANCER RESEARCH COMMUNICATIONS 2023; 3:1350-1365. [PMID: 37501683 PMCID: PMC10370362 DOI: 10.1158/2767-9764.crc-22-0373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 05/01/2023] [Accepted: 06/30/2023] [Indexed: 07/29/2023]
Abstract
Lung adenocarcinoma (LUAD) is a heterogeneous group of tumors associated with different survival rates, even when detected at an early stage. Here, we aim to investigate the biological determinants of early LUAD indolence or aggressiveness using radiomics as a surrogate of behavior. We present a set of 92 patients with LUAD with data collected across different methodologies. Patients were risk-stratified using the CT-based Score Indicative of Lung cancer Aggression (SILA) tool (0 = least aggressive, 1 = most aggressive). We grouped the patients as indolent (x ≤ 0.4, n = 14), intermediate (0.4 > x ≤ 0.6, n = 27), and aggressive (0.6 > x ≤ 1, n = 52). Using Cytometry by time of flight (CyTOF), we identified subpopulations with high HLA-DR expression that were associated with indolent behavior. In the RNA sequencing (RNA-seq) dataset, pathways related to immune response were associated with indolent behavior, while pathways associated with cell cycle and proliferation were associated with aggressive behavior. We extracted quantitative radiomics features from the CT scans of the patients. Integrating these datasets, we identified four feature signatures and four patient clusters that were associated with survival. Using single-cell RNA-seq, we found that indolent tumors had significantly more T cells and less B cells than aggressive tumors, and that the latter had a higher abundance of regulatory T cells and Th cells. In conclusion, we were able to uncover a correspondence between radiomics and tumor biology, which could improve the discrimination between indolent and aggressive LUAD tumors, enhance our knowledge in the biology of these tumors, and offer novel and personalized avenues for intervention. Significance This study provides a comprehensive profiling of LUAD indolence and aggressiveness at the biological bulk and single-cell levels, as well as at the clinical and radiomics levels. This hypothesis generating study uncovers several potential future research avenues. It also highlights the importance and power of data integration to improve our systemic understanding of LUAD and to help reduce the gap between basic science research and clinical practice.
Collapse
Affiliation(s)
- Maria-Fernanda Senosain
- Cancer Biology Graduate Program, Vanderbilt University, Nashville, Tennessee
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, Tennessee
| | - Yong Zou
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, Tennessee
| | - Khushbu Patel
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, Tennessee
| | - Shilin Zhao
- Vanderbilt Ingram Cancer Center, Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alexis Coullomb
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Dianna J. Rowe
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, Tennessee
| | - Jonathan M. Lehman
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jonathan M. Irish
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Fabien Maldonado
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael N. Kammer
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, Tennessee
| | - Vera Pancaldi
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Barcelona Supercomputing Center, Carrer de Jordi Girona, 29, 31, 08034 Barcelona, Spain
| | - Carlos F. Lopez
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee
- Department of Biomedical Informatics, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
24
|
Merk-Ahmad K, Bein J, Scharf S, Schäfer H, Bexte T, Ullrich E, Loth AG, Flinner N, Senff T, Schneider O, Hansmann ML, Piel M, Häupl B, Oellerich T, Donnadieu E, Hartmann S. The RHOA Mutation G17V Does Not Lead to Increased Migration of Human Malignant T Cells but Is Associated with Matrix Remodelling. Cancers (Basel) 2023; 15:3226. [PMID: 37370838 DOI: 10.3390/cancers15123226] [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: 05/26/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Nodal T-follicular helper cell lymphoma, angioimmunoblastic-type (AITL), is characterized by constitutional symptoms, advanced-stage disease, and generalized lymphadenopathy. A genetic hallmark of this lymphoma is the frequent occurrence of the RHOA mutation G17V in neoplastic cells, which is observed in around 60% of patients. Because RHOA is involved in both T-cell receptor downstream signalling and cell migration, we hypothesized that the characteristic presentation of AITL could be the result of enhanced tumor cell migration. Therefore, this study aimed to elucidate the impact of the RHOA variant G17V on the migration of neoplastic T cells. We transfected the T-cell lymphoma cell lines HH and HuT78 to stably express the RHOA-G17V variant. RHOA-G17V-expressing T cells did not exhibit enhanced motility compared to empty-vector-transfected cells in microchannels, a 3D collagen gel, or primary human lymphatic tissue. Cells of the HH cell line expressing RHOA-G17V had an increased number of cells with cleaved collagen compared with the empty-vector-transfected cells. Therefore, we hypothesized that the early spread of AITL tumor cells may be related to remodelling of the extracellular matrix. Accordingly, we observed a significant negative correlation between the relative area of collagen in histological sections from 18 primary AITL and the allele frequency of the RHOA-G17V mutation. In conclusion, our results suggest that the characteristic presentation of AITL with early, widespread dissemination of lymphoma cells is not the result of an enhanced migration capacity due to the RHOA-G17V mutation; instead, this feature may rather be related to extracellular matrix remodelling.
Collapse
Affiliation(s)
- Katrin Merk-Ahmad
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany
| | - Julia Bein
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany
| | - Sonja Scharf
- Frankfurt Institute for Advanced Studies, 60438 Frankfurt am Main, Germany
- Molecular Bioinformatics, Goethe University Frankfurt am Main, Robert-Mayer-Str. 11-15, 60325 Frankfurt am Main, Germany
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Hendrik Schäfer
- Frankfurt Institute for Advanced Studies, 60438 Frankfurt am Main, Germany
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Tobias Bexte
- Department for Pediatrics, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
- Experimental Immunology and Cell Therapy, Department of Pediatrics, Goethe University, 60528 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60590 Frankfurt am Main, Germany
- University Cancer Center (UCT) Frankfurt, University Hospital, Goethe University, 60590 Frankfurt am Main, Germany
| | - Evelyn Ullrich
- Department for Pediatrics, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
- Experimental Immunology and Cell Therapy, Department of Pediatrics, Goethe University, 60528 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60528 Frankfurt am Main, Germany
| | - Andreas G Loth
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Nadine Flinner
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany
| | - Tina Senff
- Institute of Pathology and Molecular Pathology, Helios Klinikum Wuppertal, 42283 Wuppertal, Germany
| | - Olga Schneider
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany
| | - Martin-Leo Hansmann
- Frankfurt Institute for Advanced Studies, 60438 Frankfurt am Main, Germany
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Matthieu Piel
- Institut Curie and Institut Pierre Gilles de Gennes, CNRS, UMR 144, PSL Research University, 75005 Paris, France
| | - Björn Häupl
- Department of Internal Medicine 2, Goethe University Hospital, 60590 Frankfurt am Main, Germany
- German Cancer Consortium, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Thomas Oellerich
- Frankfurt Cancer Institute, Goethe University, 60590 Frankfurt am Main, Germany
- University Cancer Center (UCT) Frankfurt, University Hospital, Goethe University, 60590 Frankfurt am Main, Germany
- Department of Internal Medicine 2, Goethe University Hospital, 60590 Frankfurt am Main, Germany
| | - Emmanuel Donnadieu
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, 75006 Paris, France
| | - Sylvia Hartmann
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany
| |
Collapse
|
25
|
Zhuang C, Gould JE, Enninful A, Shao S, Mak M. Biophysical and mechanobiological considerations for T-cell-based immunotherapy. Trends Pharmacol Sci 2023; 44:366-378. [PMID: 37172572 PMCID: PMC10188210 DOI: 10.1016/j.tips.2023.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 05/15/2023]
Abstract
Immunotherapies modulate the body's defense system to treat cancer. While these therapies have shown efficacy against multiple types of cancer, patient response rates are limited, and the off-target effects can be severe. Typical approaches in developing immunotherapies tend to focus on antigen targeting and molecular signaling, while overlooking biophysical and mechanobiological effects. Immune cells and tumor cells are both responsive to biophysical cues, which are prominent in the tumor microenvironment. Recent studies have shown that mechanosensing - including through Piezo1, adhesions, and Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) - influences tumor-immune interactions and immunotherapeutic efficacy. Furthermore, biophysical methods such as fluidic systems and mechanoactivation schemes can improve the controllability and manufacturing of engineered T cells, with potential for increasing therapeutic efficacy and specificity. This review focuses on leveraging advances in immune biophysics and mechanobiology toward improving chimeric antigen receptor (CAR) T-cell and anti-programmed cell death protein 1 (anti-PD-1) therapies.
Collapse
Affiliation(s)
- Chuzhi Zhuang
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Jared E Gould
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Archibald Enninful
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Stephanie Shao
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA.
| |
Collapse
|
26
|
Jiang W, Wang H, Chen W, Zhao Y, Yan B, Chen D, Dong X, Cheng J, Lin Z, Zhuo S, Wang H, Yan J. Association of collagen deep learning classifier with prognosis and chemotherapy benefits in stage II-III colon cancer. Bioeng Transl Med 2023; 8:e10526. [PMID: 37206212 PMCID: PMC10189440 DOI: 10.1002/btm2.10526] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 05/21/2023] Open
Abstract
The current tumor-node-metastasis staging system does not provide sufficient prognostic prediction or adjuvant chemotherapy benefit information for stage II-III colon cancer (CC) patients. Collagen in the tumor microenvironment affects the biological behaviors and chemotherapy response of cancer cells. Hence, in this study, we proposed a collagen deep learning (collagenDL) classifier based on the 50-layer residual network model for predicting disease-free survival (DFS) and overall survival (OS). The collagenDL classifier was significantly associated with DFS and OS (P < 0.001). The collagenDL nomogram, integrating the collagenDL classifier and three clinicopathologic predictors, improved the prediction performance, which showed satisfactory discrimination and calibration. These results were independently validated in the internal and external validation cohorts. In addition, high-risk stage II and III CC patients with high-collagenDL classifier, rather than low-collagenDL classifier, exhibited a favorable response to adjuvant chemotherapy. In conclusion, the collagenDL classifier could predict prognosis and adjuvant chemotherapy benefits in stage II-III CC patients.
Collapse
Affiliation(s)
- Wei Jiang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical MedicineSouthern Medical UniversityGuangzhouPeople's Republic of China
- School of ScienceJimei UniversityXiamenFujianPeople's Republic of China
| | - Huaiming Wang
- Department of Colorectal Surgery & Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital, Supported by National Key Clinical DisciplineSun Yat‐sen UniversityGuangzhouGuangdongPeople's Republic of China
| | - Weisheng Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical MedicineSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Yandong Zhao
- Department of Pathology, the Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongPeople's Republic of China
| | - Botao Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical MedicineSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Dexin Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical MedicineSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Xiaoyu Dong
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical MedicineSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Jiaxin Cheng
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical MedicineSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Zexi Lin
- School of ScienceJimei UniversityXiamenFujianPeople's Republic of China
| | - Shuangmu Zhuo
- School of ScienceJimei UniversityXiamenFujianPeople's Republic of China
| | - Hui Wang
- Department of Colorectal Surgery & Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital, Supported by National Key Clinical DisciplineSun Yat‐sen UniversityGuangzhouGuangdongPeople's Republic of China
| | - Jun Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical MedicineSouthern Medical UniversityGuangzhouPeople's Republic of China
| |
Collapse
|
27
|
Potashnikova DM, Saidova AA, Tvorogova AV, Anisimova AS, Botsina AY, Vasilieva EY, Margolis LB. CTLs From Patients With Atherosclerosis Show Elevated Adhesiveness and Distinct Integrin Expression Patterns on 2D Substrates. Front Med (Lausanne) 2022; 9:891916. [PMID: 35911408 PMCID: PMC9328274 DOI: 10.3389/fmed.2022.891916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/02/2022] [Indexed: 12/02/2022] Open
Abstract
Atherosclerosis is the major cause of cardiovascular disease that is characterized by plaque formation in the blood vessel wall. Atherosclerotic plaques represent sites of chronic inflammation with diverse cell content that is shifted toward the prevalence of cytotoxic T-lymphocytes (CTLs) upon plaque progression. The studies of CTL recruitment to atherosclerotic plaques require adequate in vitro models accounting for CTL interactions with chemokine-ligands and extracellular matrix fibers via surface chemokine receptors and integrins. Here we applied such a model by investigating CTL adhesion and migration on six types of coated surfaces. We assessed adhesion and motility metrics, the expression of chemokine receptors, and integrins in CTLs of patients with atherosclerosis and healthy donors. Using fibronectin, platelet-poor plasma from patients with atherosclerosis, and conditioned medium from atherosclerotic plaques we revealed the role of substrate in CTL adhesiveness: fibronectin alone and fibronectin combined with platelet-poor plasma and conditioned medium elevated the CTL adhesiveness - in patients the elevation was significantly higher than in healthy donors (p = 0.02, mixed 2-way ANOVA model). This was in line with our finding that the expression levels of integrin-coding mRNAs were elevated in the presence of fibronectin (p < 0.05) and ITGB1, ITGA1, and ITGA4 were specifically upregulated in patients compared to healthy donors (p < 0.01). Our experimental model did not affect the expression levels of mRNAs CCR4, CCR5, and CX3CR1 coding the chemokine receptors that drive T-lymphocyte migration to plaques. Thus, we demonstrated the substrate-dependence of integrin expression and discriminated CTLs from patients and healthy donors by adhesion parameters and integrin expression levels.
Collapse
Affiliation(s)
- Daria M. Potashnikova
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
- Moscow Department of Healthcare, City Clinical Hospital Named After I.V. Davydovsky, Moscow, Russia
| | - Aleena A. Saidova
- Moscow Department of Healthcare, City Clinical Hospital Named After I.V. Davydovsky, Moscow, Russia
| | - Anna V. Tvorogova
- Moscow Department of Healthcare, City Clinical Hospital Named After I.V. Davydovsky, Moscow, Russia
| | - Alexandra S. Anisimova
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
- Moscow Department of Healthcare, City Clinical Hospital Named After I.V. Davydovsky, Moscow, Russia
| | - Alexandra Yu Botsina
- Moscow Department of Healthcare, City Clinical Hospital Named After I.V. Davydovsky, Moscow, Russia
| | - Elena Yu Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
- Moscow Department of Healthcare, City Clinical Hospital Named After I.V. Davydovsky, Moscow, Russia
| | - Leonid B. Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
28
|
Nguyen DT, Ogando-Rivas E, Liu R, Wang T, Rubin J, Jin L, Tao H, Sawyer WW, Mendez-Gomez HR, Cascio M, Mitchell DA, Huang J, Sawyer WG, Sayour EJ, Castillo P. CAR T Cell Locomotion in Solid Tumor Microenvironment. Cells 2022; 11:1974. [PMID: 35741103 PMCID: PMC9221866 DOI: 10.3390/cells11121974] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 01/25/2023] Open
Abstract
The promising outcomes of chimeric antigen receptor (CAR) T cell therapy in hematologic malignancies potentiates its capability in the fight against many cancers. Nevertheless, this immunotherapy modality needs significant improvements for the treatment of solid tumors. Researchers have incrementally identified limitations and constantly pursued better CAR designs. However, even if CAR T cells are armed with optimal killer functions, they must overcome and survive suppressive barriers imposed by the tumor microenvironment (TME). In this review, we will discuss in detail the important role of TME in CAR T cell trafficking and how the intrinsic barriers contribute to an immunosuppressive phenotype and cancer progression. It is of critical importance that preclinical models can closely recapitulate the in vivo TME to better predict CAR T activity. Animal models have contributed immensely to our understanding of human diseases, but the intensive care for the animals and unreliable representation of human biology suggest in vivo models cannot be the sole approach to CAR T cell therapy. On the other hand, in vitro models for CAR T cytotoxic assessment offer valuable insights to mechanistic studies at the single cell level, but they often lack in vivo complexities, inter-individual heterogeneity, or physiologically relevant spatial dimension. Understanding the advantages and limitations of preclinical models and their applications would enable more reliable prediction of better clinical outcomes.
Collapse
Affiliation(s)
- Duy T. Nguyen
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (W.W.S.); (W.G.S.)
| | - Elizabeth Ogando-Rivas
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Ruixuan Liu
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Theodore Wang
- College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Jacob Rubin
- Warrington College of Business, University of Florida, Gainesville, FL 32610, USA;
| | - Linchun Jin
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Haipeng Tao
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - William W. Sawyer
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (W.W.S.); (W.G.S.)
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| | - Hector R. Mendez-Gomez
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Matthew Cascio
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| | - Duane A. Mitchell
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Jianping Huang
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - W. Gregory Sawyer
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (W.W.S.); (W.G.S.)
| | - Elias J. Sayour
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| | - Paul Castillo
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| |
Collapse
|
29
|
Park S, Kim H, Wang Y, Eom DS, Allard J. Zebrafish airinemes optimize their shape between ballistic and diffusive search. eLife 2022; 11:75690. [PMID: 35467525 PMCID: PMC9098217 DOI: 10.7554/elife.75690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
In addition to diffusive signals, cells in tissue also communicate via long, thin cellular protrusions, such as airinemes in zebrafish. Before establishing communication, cellular protrusions must find their target cell. Here we demonstrate that the shapes of airinemes in zebrafish are consistent with a finite persistent random walk model. The probability of contacting the target cell is maximized for a balance between ballistic search (straight) and diffusive search (highly curved, random). We find that the curvature of airinemes in zebrafish, extracted from live cell microscopy, is approximately the same value as the optimum in the simple persistent random walk model. We also explore the ability of the target cell to infer direction of the airineme's source, finding that there is a theoretical trade-off between search optimality and directional information. This provides a framework to characterize the shape, and performance objectives, of non-canonical cellular protrusions in general.
Collapse
Affiliation(s)
- Sohyeon Park
- Center for Complex Biological Systems, University of California, Irvine, Irvine, United States
| | - Hyunjoong Kim
- Department of Mathematics, University of Pennsylvania, Pennsylvania, United States
| | - Yi Wang
- Center for Complex Biological Systems, University of California, Irvine, Irvine, United States
| | - Dae Seok Eom
- Center for Complex Biological Systems, University of California, Irvine, Irvine, United States
| | - Jun Allard
- dDepartment of Physics and Astronomy, University of California, Irvine, Irvine, United States
| |
Collapse
|
30
|
Zhou X, Zhao R, Yanamandra AK, Hoth M, Qu B. Light-Sheet Scattering Microscopy to Visualize Long-Term Interactions Between Cells and Extracellular Matrix. Front Immunol 2022; 13:828634. [PMID: 35154150 PMCID: PMC8831865 DOI: 10.3389/fimmu.2022.828634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/11/2022] [Indexed: 11/16/2022] Open
Abstract
Visualizing interactions between cells and the extracellular matrix (ECM) mesh is important to understand cell behavior and regulatory mechanisms by the extracellular environment. However, long term visualization of three-dimensional (3D) matrix structures remains challenging mainly due to photobleaching or blind spots perpendicular to the imaging plane. Here, we combine label-free light-sheet scattering microcopy (LSSM) and fluorescence microscopy to solve these problems. We verified that LSSM can reliably visualize structures of collagen matrices from different origin including bovine, human and rat tail. The quality and intensity of collagen structure images acquired by LSSM did not decline with time. LSSM offers abundant wavelength choice to visualize matrix structures, maximizing combination possibilities with fluorescently-labelled cells, allowing visualizing of long-term ECM-cell interactions in 3D. Interestingly, we observed ultrathin thread-like structures between cells and matrix using LSSM, which were not observed by normal fluorescence microscopy. Transient local alignment of matrix by cell-applied forces can be observed. In summary, LSSM provides a powerful and robust approach to investigate the complex interplay between cells and ECM.
Collapse
Affiliation(s)
- Xiangda Zhou
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Renping Zhao
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Archana K Yanamandra
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany.,INM-Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Markus Hoth
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Bin Qu
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany.,INM-Leibniz Institute for New Materials, Saarbrücken, Germany
| |
Collapse
|
31
|
Abstract
Transforming growth factor-β (TGFβ) signalling controls multiple cell fate decisions during development and tissue homeostasis; hence, dysregulation of this pathway can drive several diseases, including cancer. Here we discuss the influence that TGFβ exerts on the composition and behaviour of different cell populations present in the tumour immune microenvironment, and the context-dependent functions of this cytokine in suppressing or promoting cancer. During homeostasis, TGFβ controls inflammatory responses triggered by exposure to the outside milieu in barrier tissues. Lack of TGFβ exacerbates inflammation, leading to tissue damage and cellular transformation. In contrast, as tumours progress, they leverage TGFβ to drive an unrestrained wound-healing programme in cancer-associated fibroblasts, as well as to suppress the adaptive immune system and the innate immune system. In consonance with this key role in reprogramming the tumour microenvironment, emerging data demonstrate that TGFβ-inhibitory therapies can restore cancer immunity. Indeed, this approach can synergize with other immunotherapies - including immune checkpoint blockade - to unleash robust antitumour immune responses in preclinical cancer models. Despite initial challenges in clinical translation, these findings have sparked the development of multiple therapeutic strategies that inhibit the TGFβ pathway, many of which are currently in clinical evaluation.
Collapse
Affiliation(s)
- Daniele V F Tauriello
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Elena Sancho
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain.
- Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
32
|
Rømer AMA, Thorseth ML, Madsen DH. Immune Modulatory Properties of Collagen in Cancer. Front Immunol 2021; 12:791453. [PMID: 34956223 PMCID: PMC8692250 DOI: 10.3389/fimmu.2021.791453] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022] Open
Abstract
During tumor growth the extracellular matrix (ECM) undergoes dramatic remodeling. The normal ECM is degraded and substituted with a tumor-specific ECM, which is often of higher collagen density and increased stiffness. The structure and collagen density of the tumor-specific ECM has been associated with poor prognosis in several types of cancer. However, the reason for this association is still largely unknown. Collagen can promote cancer cell growth and migration, but recent studies have shown that collagens can also affect the function and phenotype of various types of tumor-infiltrating immune cells such as tumor-associated macrophages (TAMs) and T cells. This suggests that tumor-associated collagen could have important immune modulatory functions within the tumor microenvironment, affecting cancer progression as well as the efficacy of cancer immunotherapy. The effects of tumor-associated collagen on immune cells could help explain why a high collagen density in tumors is often correlated with a poor prognosis. Knowledge about immune modulatory functions of collagen could potentially identify targets for improving current cancer therapies or for development of new treatments. In this review, the current knowledge about the ability of collagen to influence T cell activity will be summarized. This includes direct interactions with T cells as well as induction of immune suppressive activity in other immune cells such as macrophages. Additionally, the potential effects of collagen on the efficacy of cancer immunotherapy will be discussed.
Collapse
Affiliation(s)
- Anne Mette Askehøj Rømer
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Marie-Louise Thorseth
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Hargbøl Madsen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Frattolin J, Watson DJ, Bonneuil WV, Russell MJ, Fasanella Masci F, Bandara M, Brook BS, Nibbs RJB, Moore JE. The Critical Importance of Spatial and Temporal Scales in Designing and Interpreting Immune Cell Migration Assays. Cells 2021; 10:3439. [PMID: 34943947 PMCID: PMC8700135 DOI: 10.3390/cells10123439] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 02/08/2023] Open
Abstract
Intravital microscopy and other direct-imaging techniques have allowed for a characterisation of leukocyte migration that has revolutionised the field of immunology, resulting in an unprecedented understanding of the mechanisms of immune response and adaptive immunity. However, there is an assumption within the field that modern imaging techniques permit imaging parameters where the resulting cell track accurately captures a cell's motion. This notion is almost entirely untested, and the relationship between what could be observed at a given scale and the underlying cell behaviour is undefined. Insufficient spatial and temporal resolutions within migration assays can result in misrepresentation of important physiologic processes or cause subtle changes in critical cell behaviour to be missed. In this review, we contextualise how scale can affect the perceived migratory behaviour of cells, summarise the limited approaches to mitigate this effect, and establish the need for a widely implemented framework to account for scale and correct observations of cell motion. We then extend the concept of scale to new approaches that seek to bridge the current "black box" between single-cell behaviour and systemic response.
Collapse
Affiliation(s)
- Jennifer Frattolin
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (J.F.); (D.J.W.); (W.V.B.)
| | - Daniel J. Watson
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (J.F.); (D.J.W.); (W.V.B.)
| | - Willy V. Bonneuil
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (J.F.); (D.J.W.); (W.V.B.)
| | - Matthew J. Russell
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (M.J.R.); (B.S.B.)
| | - Francesca Fasanella Masci
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK; (F.F.M.); (M.B.); (R.J.B.N.)
| | - Mikaila Bandara
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK; (F.F.M.); (M.B.); (R.J.B.N.)
| | - Bindi S. Brook
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (M.J.R.); (B.S.B.)
| | - Robert J. B. Nibbs
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK; (F.F.M.); (M.B.); (R.J.B.N.)
| | - James E. Moore
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (J.F.); (D.J.W.); (W.V.B.)
| |
Collapse
|
34
|
van Steen ACI, Kempers L, Schoppmeyer R, Blokker M, Beebe DJ, Nolte MA, van Buul JD. Transendothelial migration induces differential migration dynamics of leukocytes in tissue matrix. J Cell Sci 2021; 134:272419. [PMID: 34622930 DOI: 10.1242/jcs.258690] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/27/2021] [Indexed: 01/14/2023] Open
Abstract
Leukocyte extravasation into inflamed tissue is a complex process that is difficult to capture as a whole in vitro. We employed a blood-vessel-on-a-chip model in which human endothelial cells were cultured in a tube-like lumen in a collagen-1 matrix. The vessels are leak tight, creating a barrier for molecules and leukocytes. Addition of inflammatory cytokine TNF-α (also known as TNF) caused vasoconstriction, actin remodelling and upregulation of ICAM-1. Introducing leukocytes into the vessels allowed real-time visualization of all different steps of the leukocyte transmigration cascade, including migration into the extracellular matrix. Individual cell tracking over time distinguished striking differences in migratory behaviour between T-cells and neutrophils. Neutrophils cross the endothelial layer more efficiently than T-cells, but, upon entering the matrix, neutrophils display high speed but low persistence, whereas T-cells migrate with low speed and rather linear migration. In conclusion, 3D imaging in real time of leukocyte extravasation in a vessel-on-a-chip enables detailed qualitative and quantitative analysis of different stages of the full leukocyte extravasation process in a single assay. This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Abraham C I van Steen
- Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands
| | - Lanette Kempers
- Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands
| | - Rouven Schoppmeyer
- Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.,Leeuwenhoek Centre for Advanced Microscopy (LCAM), Section of Molecular Cytology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Max Blokker
- Department of Physics, Vrije Universiteit Amsterdam, De Boelelaan 1081, 1081 HV Amsterdam, The Netherlands
| | - David J Beebe
- Department of Biomedical Engineering, Department of Pathology and Laboratory Medicine, Carbone Cancer Center, University of Wisconsin-Madison, 1111 Highland Drive, Madison, WI 53705, USA
| | - Martijn A Nolte
- Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands
| | - Jaap D van Buul
- Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.,Leeuwenhoek Centre for Advanced Microscopy (LCAM), Section of Molecular Cytology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
35
|
Zhao R, Zhou X, Khan ES, Alansary D, Friedmann KS, Yang W, Schwarz EC, del Campo A, Hoth M, Qu B. Targeting the Microtubule-Network Rescues CTL Killing Efficiency in Dense 3D Matrices. Front Immunol 2021; 12:729820. [PMID: 34484240 PMCID: PMC8416057 DOI: 10.3389/fimmu.2021.729820] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/31/2021] [Indexed: 12/11/2022] Open
Abstract
Efficacy of cytotoxic T lymphocyte (CTL)-based immunotherapy is still unsatisfactory against solid tumors, which are frequently characterized by condensed extracellular matrix. Here, using a unique 3D killing assay, we identify that the killing efficiency of primary human CTLs is substantially impaired in dense collagen matrices. Although the expression of cytotoxic proteins in CTLs remained intact in dense collagen, CTL motility was largely compromised. Using light-sheet microscopy, we found that persistence and velocity of CTL migration was influenced by the stiffness and porosity of the 3D matrix. Notably, 3D CTL velocity was strongly correlated with their nuclear deformability, which was enhanced by disruption of the microtubule network especially in dense matrices. Concomitantly, CTL migration, search efficiency, and killing efficiency in dense collagen were significantly increased in microtubule-perturbed CTLs. In addition, the chemotherapeutically used microtubule inhibitor vinblastine drastically enhanced CTL killing efficiency in dense collagen. Together, our findings suggest targeting the microtubule network as a promising strategy to enhance efficacy of CTL-based immunotherapy against solid tumors, especially stiff solid tumors.
Collapse
Affiliation(s)
- Renping Zhao
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Xiangda Zhou
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Essak S. Khan
- INM-Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Dalia Alansary
- Molecular Biophysics, CIPMM, School of Medicine, Saarland University, Homburg, Germany
| | - Kim S. Friedmann
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Wenjuan Yang
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Eva C. Schwarz
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | | | - Markus Hoth
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Bin Qu
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
- INM-Leibniz Institute for New Materials, Saarbrücken, Germany
| |
Collapse
|
36
|
Wortel IMN, Niculescu I, Kolijn PM, Gov NS, de Boer RJ, Textor J. Local actin dynamics couple speed and persistence in a cellular Potts model of cell migration. Biophys J 2021; 120:2609-2622. [PMID: 34022237 PMCID: PMC8390880 DOI: 10.1016/j.bpj.2021.04.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 03/24/2021] [Accepted: 04/14/2021] [Indexed: 12/28/2022] Open
Abstract
Cell migration is astoundingly diverse. Molecular signatures, cell-cell interactions, and environmental structures each play their part in shaping cell motion, yielding numerous morphologies and migration modes. Nevertheless, in recent years, a simple unifying law was found to describe cell migration across many different cell types and contexts: faster cells turn less frequently. This universal coupling between speed and persistence (UCSP) was explained by retrograde actin flow from front to back, but it remains unclear how this mechanism generalizes to cells with complex shapes and cells migrating in structured environments, which may not have a well-defined front-to-back orientation. Here, we present an in-depth characterization of an existing cellular Potts model, in which cells polarize dynamically from a combination of local actin dynamics (stimulating protrusions) and global membrane tension along the perimeter (inhibiting protrusions). We first show that the UCSP emerges spontaneously in this model through a cross talk of intracellular mechanisms, cell shape, and environmental constraints, resembling the dynamic nature of cell migration in vivo. Importantly, we find that local protrusion dynamics suffice to reproduce the UCSP-even in cases in which no clear global, front-to-back polarity exists. We then harness the spatial nature of the cellular Potts model to show how cell shape dynamics limit both the speed and persistence a cell can reach and how a rigid environment such as the skin can restrict cell motility even further. Our results broaden the range of potential mechanisms underlying the speed-persistence coupling that has emerged as a fundamental property of migrating cells.
Collapse
Affiliation(s)
- Inge M N Wortel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands; Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, the Netherlands.
| | - Ioana Niculescu
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht University, Utrecht, the Netherlands
| | - P Martijn Kolijn
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht University, Utrecht, the Netherlands
| | - Nir S Gov
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot, Israel
| | - Rob J de Boer
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht University, Utrecht, the Netherlands
| | - Johannes Textor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands; Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, the Netherlands.
| |
Collapse
|
37
|
Zhang K, Feng Q, Fang Z, Gu L, Bian L. Structurally Dynamic Hydrogels for Biomedical Applications: Pursuing a Fine Balance between Macroscopic Stability and Microscopic Dynamics. Chem Rev 2021; 121:11149-11193. [PMID: 34189903 DOI: 10.1021/acs.chemrev.1c00071] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Owing to their unique chemical and physical properties, hydrogels are attracting increasing attention in both basic and translational biomedical studies. Although the classical hydrogels with static networks have been widely reported for decades, a growing number of recent studies have shown that structurally dynamic hydrogels can better mimic the dynamics and functions of natural extracellular matrix (ECM) in soft tissues. These synthetic materials with defined compositions can recapitulate key chemical and biophysical properties of living tissues, providing an important means to understanding the mechanisms by which cells sense and remodel their surrounding microenvironments. This review begins with the overall expectation and design principles of dynamic hydrogels. We then highlight recent progress in the fabrication strategies of dynamic hydrogels including both degradation-dependent and degradation-independent approaches, followed by their unique properties and use in biomedical applications such as regenerative medicine, drug delivery, and 3D culture. Finally, challenges and emerging trends in the development and application of dynamic hydrogels are discussed.
Collapse
Affiliation(s)
- Kunyu Zhang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Qian Feng
- Bioengineering College, Chongqing University, Chongqing 400044, People's Republic of China
| | - Zhiwei Fang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Luo Gu
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, People's Republic of China.,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, People's Republic of China.,Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, People's Republic of China.,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, People's Republic of China.,Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, People's Republic of China
| |
Collapse
|
38
|
Vesperini D, Montalvo G, Qu B, Lautenschläger F. Characterization of immune cell migration using microfabrication. Biophys Rev 2021; 13:185-202. [PMID: 34290841 PMCID: PMC8285443 DOI: 10.1007/s12551-021-00787-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
The immune system provides our defense against pathogens and aberrant cells, including tumorigenic and infected cells. Motility is one of the fundamental characteristics that enable immune cells to find invading pathogens, control tissue damage, and eliminate primary developing tumors, even in the absence of external treatments. These processes are termed "immune surveillance." Migration disorders of immune cells are related to autoimmune diseases, chronic inflammation, and tumor evasion. It is therefore essential to characterize immune cell motility in different physiologically and pathologically relevant scenarios to understand the regulatory mechanisms of functionality of immune responses. This review is focused on immune cell migration, to define the underlying mechanisms and the corresponding investigative approaches. We highlight the challenges that immune cells encounter in vivo, and the microfabrication methods to mimic particular aspects of their microenvironment. We discuss the advantages and disadvantages of the proposed tools, and provide information on how to access them. Furthermore, we summarize the directional cues that regulate individual immune cell migration, and discuss the behavior of immune cells in a complex environment composed of multiple directional cues.
Collapse
Affiliation(s)
- Doriane Vesperini
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany
- Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
| | - Galia Montalvo
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany
- Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Bin Qu
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, 66421 Homburg, Germany
- Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| | - Franziska Lautenschläger
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany
- Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
| |
Collapse
|
39
|
Gordon-Weeks A, Yuzhalin AE. Cancer Extracellular Matrix Proteins Regulate Tumour Immunity. Cancers (Basel) 2020; 12:E3331. [PMID: 33187209 PMCID: PMC7696558 DOI: 10.3390/cancers12113331] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
The extracellular matrix (ECM) plays an increasingly recognised role in the development and progression of cancer. Whilst significant progress has been made in targeting aspects of the tumour microenvironment such as tumour immunity and angiogenesis, there are no therapies that address the cancer ECM. Importantly, immune function relies heavily on the structure, physics and composition of the ECM, indicating that cancer ECM and immunity are mechanistically inseparable. In this review we highlight mechanisms by which the ECM shapes tumour immunity, identifying potential therapeutic targets within the ECM. These data indicate that to fully realise the potential of cancer immunotherapy, the cancer ECM requires simultaneous consideration.
Collapse
Affiliation(s)
- Alex Gordon-Weeks
- Nuffield Department of Surgical Sciences, University of Oxford, Room 6607, Level 6 John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Arseniy E. Yuzhalin
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| |
Collapse
|