1
|
Kibet M, Abebayehu D. Crosstalk between T cells and fibroblasts in biomaterial-mediated fibrosis. Matrix Biol Plus 2025; 26:100172. [PMID: 40226302 PMCID: PMC11986236 DOI: 10.1016/j.mbplus.2025.100172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
Biomaterial implants are a critical aspect of our medical therapies and biomedical research and come in various forms: stents, implantable glucose sensors, orthopedic implants, silicone implants, drug delivery systems, and tissue engineered scaffolds. Their implantation triggers a series of biological responses that often times lead to the foreign body response and subsequent fibrotic encapsulation, a dense ECM-rich capsule that isolates the biomaterial and renders it ineffective. These responses lead to the failure of biomaterials and is a major hurdle to overcome and in promoting their success. Much attention has been given to macrophage populations for the inflammatory component of these responses to biomaterials but recent work has identified an important role of T cells and their ability to modulate fibroblast activity and vice versa. In this review, we focus on T cell-fibroblast crosstalk by exploring T cell subsets, critical signaling pathways, and fibroblast populations that have been shown to dictate biomaterial-mediated fibrosis. We then highlight emerging technologies and model systems that enable new insights and avenues to T cell-fibroblast crosstalk that will improve biomaterial outcomes.
Collapse
Affiliation(s)
- Mathew Kibet
- Department of Biomedical Engineering, School of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, United States
| | - Daniel Abebayehu
- Department of Biomedical Engineering, School of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, United States
| |
Collapse
|
2
|
Yassin AAK, Ureña Martin C, Le Saux G, Pandey A, Tzadka S, Toledo E, Pandit JJ, Sherf T, Nusbaum I, Bhattachrya B, Banerji R, Greenshpan Y, Abu Ahmad MA, Radinsky O, Sklartz M, Gazit R, Elkabets M, Ghassemi S, Cohen O, Schvartzman M, Porgador A. Mechanostimulatory Platform for Improved CAR T Cell Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2412482. [PMID: 40348587 DOI: 10.1002/adma.202412482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 04/29/2025] [Indexed: 05/14/2025]
Abstract
Chimeric Antigen Receptor T (CAR T) cell immunotherapy has revolutionized cancer treatment, yet it is hindered by rapid T-cell exhaustion caused by uncontrolled activation during CAR generation. Leveraging insights into T-cell mechanosensing, a novel mechanostimulatory platform is engineered for T-cell activation based on an antigen-carrying surface with controlled elasticity and nanotopography. The platform is designed to optimize and balance T-cell exhaustion, proliferation, and CAR expression. It enhances the differentiation of T cells into the central memory subset, which is crucial for the persistence of CAR T cell therapy's anticancer effects. The platform produces CAR T cells with higher antitumor efficacy, as validated through ex vivo experiments, and with higher in vivo persistence and ability to suppress tumor proliferation, as compared to CAR T cells generated by standard protocols. RNA-seq analysis confirmed an increased transcriptional signature of central memory T cells. Furthermore, this platform completely eliminates T-cell toxicity associated with the non-viral transfection process typically observed with standard activation methods. This platform presents a promising pathway for improving the efficiency and safety of CAR T cell therapy.
Collapse
Affiliation(s)
- Abed Al-Kader Yassin
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Carlos Ureña Martin
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Guillaume Le Saux
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Ashish Pandey
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Sivan Tzadka
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Esti Toledo
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Jatin Jawhir Pandit
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Tomer Sherf
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Idan Nusbaum
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Baisali Bhattachrya
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Rajashri Banerji
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Yariv Greenshpan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Muhammad Abu Abu Ahmad
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Olga Radinsky
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Menachem Sklartz
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Roi Gazit
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Sabah Ghassemi
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ofir Cohen
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Department of Software and Information System Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Mark Schvartzman
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| |
Collapse
|
3
|
Minopoli A, Perini G, Cui L, Palmieri V, De Spirito M, Papi M. Biomaterial-Driven 3D Scaffolds for Immune Cell Expansion toward Personalized Immunotherapy. Acta Biomater 2025:S1742-7061(25)00351-4. [PMID: 40348072 DOI: 10.1016/j.actbio.2025.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 04/12/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Immunotherapy has emerged as a transformative medical approach in recent years, providing novel treatments for cancer eradication, autoimmune disorders, and infectious diseases. Fundamental to the success of therapy is the enrichment of the immune cell population, particularly T cells, natural killer cells, and dendritic cells. However, achieving a robust and long-term proliferation of immune cells is still challenging both in vivo and ex vivo. In vivo expansion leverages the patient's natural microenvironment and regulatory mechanisms through therapeutic interventions like immune checkpoint inhibitors, cytokine therapy, and targeted antibodies. This approach fosters long-term immune memory and sustained protection. In contrast, ex vivo expansion involves isolation, manipulation, and expansion of the immune cells under controlled conditions before reinfusion, allowing for precise control over the process and generating potent immune cell populations. Hydrogels, due to their tunable biomechanical properties, high biocompatibility, and ability to mimic the extracellular matrix, provide an ideal platform for both in vivo and ex vivo immune cell expansion. For instance, hydrogel-based scaffolds or beads can facilitate a controlled and efficient expansion of immune cells ex vivo, whereas injectable and implantable hydrogels can provide innovative solutions for enhancing immune cell activity within the patient supporting prolonged immune cell activity. This review aims to elucidate the importance of hydrogel-based strategies in immune cell expansion, advancing the development of effective, personalized immunotherapies to improve patient outcomes. STATEMENT OF SIGNIFICANCE: This review highlights the transformative potential of hydrogel-based 3D scaffolds in advancing personalized immunotherapy. By integrating in vivo and ex vivo strategies, hydrogels provide an innovative platform to enhance immune cell expansion, addressing critical challenges in immunotherapy. The discussion emphasizes the unique biomechanical and biochemical tunability of hydrogels, enabling precise mimicry of the extracellular matrix to support T cell proliferation, activation, and memory formation. These advances offer scalable, cost-effective solutions for producing high-quality immune cells, contributing to more effective cancer treatments, autoimmune disease management, and infectious disease control. By bridging materials science and immunology, this work underscores the pivotal role of hydrogels in shaping the future of immune-based therapies.
Collapse
Affiliation(s)
- Antonio Minopoli
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168, Rome, Italy
| | - Giordano Perini
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168, Rome, Italy
| | - Lishan Cui
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168, Rome, Italy
| | - Valentina Palmieri
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168, Rome, Italy; Istituto dei Sistemi Complessi, Consiglio Nazionale delle Ricerche, CNR, via dei Taurini 19, 00185 Rome, Italy
| | - Marco De Spirito
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168, Rome, Italy.
| | - Massimiliano Papi
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168, Rome, Italy.
| |
Collapse
|
4
|
Lei M, Chen G. Integration of mechanics and immunology: Perspective for understanding fibrotic disease mechanisms and innovating therapeutic strategies. Acta Biomater 2025:S1742-7061(25)00333-2. [PMID: 40324516 DOI: 10.1016/j.actbio.2025.05.011] [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: 01/27/2025] [Revised: 05/01/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
The treatment of fibrotic diseases has long posed a medical challenge due to the complex mechanisms underlying their occurrence and progression. Emerging evidence suggests that fibrosis development is influenced not only by biochemical factors but also by the activation of mechanotransduction in response to mechanical stimuli. Mechanoimmunology, an interdisciplinary field that examines how the immune system is influenced by physical forces and mechanical environments, has recently demonstrated significant importance and considerable potential for application in the study of fibrotic diseases. While the mechanisms by which biochemical signals regulate the immune system have been extensively explored, the progression of fibrosis is often impacted by both immune dysregulation and mechanical changes. During fibrosis, immune cells encounter strong mechanical stimuli, such as stiffer substrates and altered viscoelasticity, which activate their own mechanotransduction pathways and subsequently influence fibrosis progression. Targeting the mechanosensation of immune cells to enhance or inhibit their mechanoreception and mechanotransduction, thereby enhancing the anti-fibrotic role they play in the fibrotic process, could help innovate therapeutic strategies for fibrotic diseases. STATEMENT OF SIGNIFICANCE: Fibrotic disease progression is often associated with dysregulation of both tissue mechanical properties and immune responses. The fibrotic microenvironment's altered mechanical properties both result from and drive fibrosis, while immune cells actively sense and respond to these mechanical cues through mechanotransduction pathways. Emerging mechanoimmunology research highlights how mechanical stimuli influence immune cell behavior, yet the precise regulatory mechanisms remain unclear. This review examines mechanical communication in fibrosis, focusing on immune cells' mechanosensing capabilities and their role in disease progression, which helps to enhance our understanding of the pathogenesis of fibrosis and inform innovative strategies to open up mechano-immune pathways targeting fibrosis therapy.
Collapse
Affiliation(s)
- Min Lei
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Guobao Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China.
| |
Collapse
|
5
|
Sanati M, Pieterman I, Levy N, Akbari T, Tavakoli M, Hassani Najafabadi A, Amin Yavari S. Osteoimmunomodulation by bone implant materials: harnessing physicochemical properties and chemical composition. Biomater Sci 2025. [PMID: 40289736 DOI: 10.1039/d5bm00357a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Chronic inflammation at bone defect sites can impede regenerative processes, but local immune responses can be adjusted to promote healing. Regulating the osteoimmune microenvironment, particularly through macrophage polarization, has become a key focus in bone regeneration research. While bone implants are crucial for addressing significant bone defects, they are often recognized by the immune system as foreign, triggering inflammation that leads to bone resorption and implant issues like fibrous encapsulation and aseptic loosening. Developing osteoimmunomodulatory implants offers a promising approach to transforming destructive inflammation into healing processes, enhancing implant integration and bone regeneration. This review explores strategies based on tuning the physicochemical attributes and chemical composition of materials in engineering osteoimmunomodulatory and pro-regenerative bone implants.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Ines Pieterman
- Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Natacha Levy
- Metabolic Diseases Pediatrics Division, University Medical Centre Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tayebeh Akbari
- Department of Microbiology, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Mohamadreza Tavakoli
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands.
- Regenerative Medicine Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
6
|
Pandit JJ, Yassin AAK, Martin CU, Saux GL, Porgador A, Schvartzman M. Effect of binary mechanical environment on T cell function. Acta Biomater 2025; 195:83-93. [PMID: 39952341 DOI: 10.1016/j.actbio.2025.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
T cells, key players in the immune system, recognize antigens via T-cell receptors (TCRs) and require additional costimulatory and cytokine signals for full activation. Beyond biochemical signals, T cells also respond to mechanical cues such as tissue stiffness. Traditional ex-vivo mechanostimulating platforms, however, present a uniform mechanical environment, unlike the heterogeneous conditions T cells encounter in-vivo. This work introduces a mechanically-biphasic T-cell stimulating surface, with alternating soft and stiff microdomains, to mimic the complex mechanical signals T cells face. Results show that T cells exposed to this biphasic environment do not average the mechanical signals but instead respond similarly to those on a homogeneously soft surface, leading to lower activation compared to those on a stiff surface. Interestingly, long-term exposure to these patterns enhances the proliferation of central memory and effector T cell phenotypes, similar to stiff environments. These findings reveal the non-linear nature of T cell mechanosensing and suggest that mechanical heterogeneity plays a critical role in modulating T cell responses, providing new insights into T cell activation and potential implications for immunotherapies. STATEMENT OF SIGNIFICANCE: This research offers a fresh perspective in T cell mehanosensing, an important yet underexplored aspect of immunity. While previous studies have demonstrated that T cells sense homogeneous mechanical environments ex-vivo, their ability to discern and respond to simultaneous mechanical cues-resembling the complexity of in-vivo conditions-remained unexamined. By designing a mechanically patterned surface with alternating soft and stiff microdomains, this study simulates the diverse mechanical landscape encountered by T cells in-vivo. The findings reveal that T cells predominantly respond to this pattern as they would to a uniformly soft environment. This insight, showing that mechanical signals shape T cell activation and promote specific phenotypes, enhances our understanding of T cell biology and points to new directions for immunotherapy development.
Collapse
Affiliation(s)
- Jatin Jawhir Pandit
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva , Israel; Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva , Israel
| | - Abed Al-Kader Yassin
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Carlos Ureña Martin
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva , Israel; Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva , Israel
| | - Guillaume Le Saux
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva , Israel; Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva , Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Mark Schvartzman
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva , Israel; Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva , Israel.
| |
Collapse
|
7
|
Mei J, Yang K, Zhang X, Luo Z, Tian M, Fan H, Chu J, Zhang Y, Ding J, Xu J, Cai Y, Yin Y. Intratumoral Collagen Deposition Supports Angiogenesis Suggesting Anti-angiogenic Therapy in Armored and Cold Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409147. [PMID: 39823457 PMCID: PMC11904994 DOI: 10.1002/advs.202409147] [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: 08/20/2024] [Revised: 01/05/2025] [Indexed: 01/19/2025]
Abstract
A previous study classifies solid tumors based on collagen deposition and immune infiltration abundance, identifying a refractory subtype termed armored & cold tumors, characterized by elevated collagen deposition and diminished immune infiltration. Beyond its impact on immune infiltration, collagen deposition also influences tumor angiogenesis. This study systematically analyzes the association between immuno-collagenic subtypes and angiogenesis across diverse cancer types. As a result, armored & cold tumors exhibit the highest angiogenic activity in lung adenocarcinoma (LUAD). Single-cell and spatial transcriptomics reveal close interactions and spatial co-localization of fibroblasts and endothelial cells. In vitro experiments demonstrate that collagen stimulates tumor cells to express vascular endothelial growth factor A (VEGFA) and directly enhances vessel formation and endothelial cell proliferation through sex determining region Y box 18 (SOX18) upregulation. Collagen inhibition via multiple approaches effectively suppresses tumor angiogenesis in vivo. In addition, armored & cold tumors display superior responsiveness to anti-angiogenic therapy in advanced LUAD cohorts. Post-immunotherapy resistance, the transformation into armored & cold tumors emerges as a potential biomarker for selecting anti-angiogenic therapy. In summary, collagen deposition is shown to drive angiogenesis across various cancers, providing a novel and actionable framework to refine therapeutic strategies combining chemotherapy with anti-angiogenic treatments.
Collapse
Affiliation(s)
- Jie Mei
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Kai Yang
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Xinkang Zhang
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Zhiwen Luo
- Department of Sports MedicineHuashan Hospital Affiliated to Fudan UniversityShanghai200040P. R. China
| | - Min Tian
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Hanfang Fan
- Departments of OncologyWuxi People's HospitalThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023P. R. China
| | - Jiahui Chu
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Yan Zhang
- Departments of GynecologyThe Women's Hospital Affiliated to Jiangnan UniversityWuxi214023China
| | - Junli Ding
- Departments of OncologyWuxi People's HospitalThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023P. R. China
| | - Junying Xu
- Departments of OncologyWuxi People's HospitalThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023P. R. China
| | - Yun Cai
- Central LaboratoryChangzhou Jintan First People's HospitalThe Affiliated Jintan Hospital of Jiangsu UniversityChangzhouJiangsu213200P. R. China
| | - Yongmei Yin
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- Jiangsu Key Lab of Cancer BiomarkersPrevention and TreatmentCollaborative Innovation Center for Personalized Cancer MedicineNanjing Medical UniversityNanjingJiangsuP. R. China
| |
Collapse
|
8
|
Song J, Lu Y, Liu L, Han X, Meng Y, Heng BC, Zhang X, Cui Q, Liu Z, Guo Y, Zheng X, You F, Lu D, Zhang X, Deng X. Charged substrate treatment enhances T cell mediated cancer immunotherapy. Nat Commun 2025; 16:1585. [PMID: 39939595 PMCID: PMC11821856 DOI: 10.1038/s41467-025-56858-y] [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: 05/03/2024] [Accepted: 01/27/2025] [Indexed: 02/14/2025] Open
Abstract
Biophysical cues play a crucial role in T cell biology, yet their implications in adoptive T cell therapy (ACT) remain largely unknown. Here, we investigate the effect of electrical stimuli on CD8+ T cells using a charged substrate composed of electroactive nanocomposites with tunable surface charge intensities. Electrical stimuli enhance the persistence and tumor-suppressive efficacy of transferred T cells, with effects dependent on substrate charge. Single-cell RNA-sequencing analysis unveils a decrease in virtual memory T (Tvm) cells and an increase in proliferative potential T (Tpp) cells, which exhibit superior antitumor activity and metabolic adaptations relative to those treated with uncharged substrate. ATAC-seq profiling demonstrates heightened accessibility at upstream binding sites for EGR1, a transcription factor critical for Tpp cell differentiation. Mechanistically, the charged substrate disrupts ionic TCR-lipid interactions, amplifies TCR signaling, and activates EGR1, thereby impeding Tvm polarization during ex vivo culture. Our findings thus highlight the importance of extracellular electrical stimuli in shaping T cell fate, offering potential for optimizing ACT for therapeutic applications.
Collapse
Affiliation(s)
- Jia Song
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Yanhui Lu
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Lulu Liu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Xiaoyu Han
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Yanhong Meng
- Department of Clinical Laboratory, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Boon Chin Heng
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Xin Zhang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, PR China
| | - Qun Cui
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Ziqi Liu
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Yusi Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Xiaona Zheng
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Fuping You
- Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, PR China
| | - Dan Lu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, PR China.
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China.
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China.
- Oral Translational Medicine Research Center, Joint Training base for Shanxi Provincial Key Laboratory in Oral and Maxillofacial Repair, Reconstruction and Regeneration, The First People's Hospital of Jinzhong, Jinzhong, Shanxi Province, PR China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China.
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China.
| |
Collapse
|
9
|
Schultheiss PJ, Pulkundwar A, Li W, Kam LC. Taming Variability in T-Cell Mechanosensing. Cells 2025; 14:203. [PMID: 39936994 PMCID: PMC11817355 DOI: 10.3390/cells14030203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/13/2025] Open
Abstract
A central step in T-cell immunotherapy is the expansion of a starting population into therapeutically potent numbers of these "living drugs". This process can be enhanced by replacing the mechanically stiff materials used for activation with softer counterparts. However, this mechanosensitive expansion response varies between individuals, impeding the full deployment of potential cell immunotherapy. This report identifies the sources of this variability, ultimately improving the reliability of T-cell expansion. T cells from a cohort of healthy donors were phenotypically characterized, activated, and expanded in vitro on soft and hard substrates, capturing and quantifying a wide range of mechanosensing responses. An analysis of expansion against demographic and phenotypic features correlated mechanosensing with the percentage of effector T cells (TEffs) in the starting population. Depletion experiments confirmed that TEffs mediate mechanosensitive expansion but also suggest that these cells are not responsible for large-scale cell production. Instead, population-level expansion results from interactions between T-cell subtypes. By providing a framework and experimental approach to understanding donor variability, the results of this study will improve the success and reliability of T-cell immunotherapy.
Collapse
Affiliation(s)
- Paula J. Schultheiss
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA; (P.J.S.); (A.P.)
| | - Aarya Pulkundwar
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA; (P.J.S.); (A.P.)
| | - Wangqi Li
- Department of Computer Science, Columbia University, New York, NY 10027, USA;
| | - Lance C. Kam
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA; (P.J.S.); (A.P.)
- Department of Medicine, Columbia University, New York, NY 10027, USA
| |
Collapse
|
10
|
Du H, Rose JP, Bons J, Guo L, Valentino TR, Wu F, Burton JB, Basisty N, Manwaring-Mueller M, Makhijani P, Chen N, Chang V, Winer S, Campisi J, Furman D, Nagy A, Schilling B, Winer DA. Substrate stiffness dictates unique doxorubicin-induced senescence-associated secretory phenotypes and transcriptomic signatures in human pulmonary fibroblasts. GeroScience 2025:10.1007/s11357-025-01507-x. [PMID: 39826027 DOI: 10.1007/s11357-025-01507-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 12/31/2024] [Indexed: 01/20/2025] Open
Abstract
Cells are subjected to dynamic mechanical environments which impart forces and induce cellular responses. In age-related conditions like pulmonary fibrosis, there is both an increase in tissue stiffness and an accumulation of senescent cells. While senescent cells produce a senescence-associated secretory phenotype (SASP), the impact of physical stimuli on both cellular senescence and the SASP is not well understood. Here, we show that mechanical tension, modeled using cell culture substrate rigidity, influences senescent cell markers like SA-β-gal and secretory phenotypes. Comparing human primary pulmonary fibroblasts (IMR-90) cultured on physiological (2 kPa), fibrotic (50 kPa), and plastic (approximately 3 GPa) substrates, followed by senescence induction using doxorubicin, we identified unique high-stiffness-driven secretory protein profiles using mass spectrometry and transcriptomic signatures, both showing an enrichment in collagen proteins. Consistently, clusters of p21 + cells are seen in fibrotic regions of bleomycin induced pulmonary fibrosis in mice. Computational meta-analysis of single-cell RNA sequencing datasets from human interstitial lung disease confirmed these stiffness SASP genes are highly expressed in disease fibroblasts and strongly correlate with mechanotransduction and senescence-related pathways. Thus, mechanical forces shape cell senescence and their secretory phenotypes.
Collapse
Affiliation(s)
- Huixun Du
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Jacob P Rose
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Joanna Bons
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Li Guo
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | | | - Fei Wu
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Jordan B Burton
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Nathan Basisty
- Longitudinal Studies Section, Translational Gerontology Branch, NIA, NIH, Baltimore, MA, USA
| | | | - Priya Makhijani
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Nan Chen
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Veronica Chang
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Shawn Winer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, , Canada
| | - Judith Campisi
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - David Furman
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Andras Nagy
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Birgit Schilling
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Daniel A Winer
- Buck Institute for Research On Aging, Novato, CA, 94945, USA.
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, M5G 1L7, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
11
|
Kang C, Chen P, Yi X, Li D, Hu Y, Yang Y, Cai H, Li B, Wu C. Amoeboid cells undergo durotaxis with soft end polarized NMIIA. eLife 2024; 13:RP96821. [PMID: 39671466 PMCID: PMC11643633 DOI: 10.7554/elife.96821] [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] [Indexed: 12/15/2024] Open
Abstract
Cell migration towards stiff substrates has been coined as durotaxis and implicated in development, wound healing, and cancer, where complex interplays between immune and non-immune cells are present. Compared to the emerging mechanisms underlying the strongly adhesive mesenchymal durotaxis, little is known about whether immune cells - migrating in amoeboid mode - could follow mechanical cues. Here, we develop an imaging-based confined migration device with a stiffness gradient. By tracking live cell trajectory and analyzing the directionality of T cells and neutrophils, we observe that amoeboid cells can durotax. We further delineate the underlying mechanism to involve non-muscle myosin IIA (NMIIA) polarization towards the soft-matrix-side but may not require differential actin flow up- or down-stiffness gradient. Using the protista Dictyostelium, we demonstrate the evolutionary conservation of amoeboid durotaxis. Finally, these experimental phenomena are theoretically captured by an active gel model capable of mechanosensing. Collectively, these results may shed new lights on immune surveillance and recently identified confined migration of cancer cells, within the mechanically inhomogeneous tumor microenvironment or the inflamed fibrotic tissues.
Collapse
Affiliation(s)
- Chenlu Kang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science CenterBeijingChina
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular OncologyBeijingChina
- International Cancer Institute, Peking UniversityBeijingChina
| | - Pengcheng Chen
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua UniversityBeijingChina
| | - Xin Yi
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science CenterBeijingChina
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular OncologyBeijingChina
- International Cancer Institute, Peking UniversityBeijingChina
| | - Dong Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of SciencesBeijingChina
- School of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Yiping Hu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science CenterBeijingChina
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular OncologyBeijingChina
- International Cancer Institute, Peking UniversityBeijingChina
| | - Yihong Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of SciencesBeijingChina
- College of Life Sciences, University of Chinese Academy of SciencesBeijingChina
| | - Huaqing Cai
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of SciencesBeijingChina
- College of Life Sciences, University of Chinese Academy of SciencesBeijingChina
| | - Bo Li
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua UniversityBeijingChina
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science CenterBeijingChina
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular OncologyBeijingChina
- International Cancer Institute, Peking UniversityBeijingChina
| |
Collapse
|
12
|
Feng X, Cao F, Wu X, Xie W, Wang P, Jiang H. Targeting extracellular matrix stiffness for cancer therapy. Front Immunol 2024; 15:1467602. [PMID: 39697341 PMCID: PMC11653020 DOI: 10.3389/fimmu.2024.1467602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/06/2024] [Indexed: 12/20/2024] Open
Abstract
The physical characteristics of the tumor microenvironment (TME) include solid stress, interstitial fluid pressure, tissue stiffness and microarchitecture. Among them, abnormal changes in tissue stiffness hinder drug delivery, inhibit infiltration of immune killer cells to the tumor site, and contribute to tumor resistance to immunotherapy. Therefore, targeting tissue stiffness to increase the infiltration of drugs and immune cells can offer a powerful support and opportunities to improve the immunotherapy efficacy in solid tumors. In this review, we discuss the mechanical properties of tumors, the impact of a stiff TME on tumor cells and immune cells, and the strategies to modulate tumor mechanics.
Collapse
Affiliation(s)
- Xiuqin Feng
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fujun Cao
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiangji Wu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenyan Xie
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Jiang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Liu Z, Li YR, Yang Y, Zhu Y, Yuan W, Hoffman T, Wu Y, Zhu E, Zarubova J, Shen J, Nan H, Yeh KW, Hasani-Sadrabadi MM, Zhu Y, Fang Y, Ge X, Li Z, Soto J, Hsiai T, Yang L, Li S. Viscoelastic synthetic antigen-presenting cells for augmenting the potency of cancer therapies. Nat Biomed Eng 2024; 8:1615-1633. [PMID: 39455719 DOI: 10.1038/s41551-024-01272-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 09/26/2024] [Indexed: 10/28/2024]
Abstract
The use of synthetic antigen-presenting cells to activate and expand engineered T cells for the treatment of cancers typically results in therapies that are suboptimal in effectiveness and durability. Here we describe a high-throughput microfluidic system for the fabrication of synthetic cells mimicking the viscoelastic and T-cell-activation properties of antigen-presenting cells. Compared with rigid or elastic microspheres, the synthetic viscoelastic T-cell-activating cells (SynVACs) led to substantial enhancements in the expansion of human CD8+ T cells and to the suppression of the formation of regulatory T cells. Notably, activating and expanding chimaeric antigen receptor (CAR) T cells with SynVACs led to a CAR-transduction efficiency of approximately 90% and to substantial increases in T memory stem cells. The engineered CAR T cells eliminated tumour cells in a mouse model of human lymphoma, suppressed tumour growth in mice with human ovarian cancer xenografts, persisted for longer periods and reduced tumour-recurrence risk. Our findings underscore the crucial roles of viscoelasticity in T-cell engineering and highlight the utility of SynVACs in cancer therapy.
Collapse
Affiliation(s)
- Zeyang Liu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yan-Ruide Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Youcheng Yang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yu Zhu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Weihao Yuan
- Section of Restorative Dentistry, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Tyler Hoffman
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yifan Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Enbo Zhu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jana Zarubova
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jun Shen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Haochen Nan
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kun-Wei Yeh
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Yichen Zhu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ying Fang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xinyang Ge
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zhizhong Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jennifer Soto
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tzung Hsiai
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
14
|
Lee CJ, Jang TY, Jeon SE, Yun HJ, Cho YH, Lim DY, Nam JS. The dysadherin/MMP9 axis modifies the extracellular matrix to accelerate colorectal cancer progression. Nat Commun 2024; 15:10422. [PMID: 39613801 DOI: 10.1038/s41467-024-54920-9] [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: 02/12/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024] Open
Abstract
The dynamic alteration of the tumor microenvironment (TME) serves as a driving force behind the progression and metastasis of colorectal cancer (CRC). Within the intricate TME, a pivotal player is the extracellular matrix (ECM), where modifications in components, degradation, and stiffness are considered critical factors in tumor development. In this study, we find that the membrane glycoprotein dysadherin directly targets matrix metalloprotease 9 (MMP9), initiating ECM remodeling within the TME and amplifying cancer progression. Mechanistically, the dysadherin/MMP9 axis not only enhances CRC cell invasiveness and ECM proteolytic activity but also activates cancer-associated fibroblasts, orchestrating the restructuring of the ECM through the synthesis of its components in human CRC cells, patient samples, and mouse models. Notably, disruption of ECM reorganization by dysadherin knockout results in a discernible reduction in the immunosuppressive and proangiogenic milieu in a humanized mouse model. Intriguingly, these effects are reversed upon the overexpression of MMP9, highlighting the intricate and pivotal role of the dysadherin/MMP9 axis in shaping the development of a malignant TME. Therefore, our findings not only highlight that dysadherin contributes to CRC progression by influencing the TME through ECM remodeling but also suggest that dysadherin may be a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Choong-Jae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Tae-Young Jang
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - So-El Jeon
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Hyeon-Ji Yun
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Yeong-Hoon Cho
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Da-Ye Lim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jeong-Seok Nam
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
15
|
Du H, Rose JP, Bons J, Guo L, Valentino TR, Wu F, Burton JB, Basisty N, Manwaring-Mueller M, Makhijani P, Chen N, Chang V, Winer S, Campisi J, Furman D, Nagy A, Schilling B, Winer DA. Substrate Stiffness Dictates Unique Doxorubicin-induced Senescence-associated Secretory Phenotypes and Transcriptomic Signatures in Human Pulmonary Fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.623471. [PMID: 39605579 PMCID: PMC11601487 DOI: 10.1101/2024.11.18.623471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Cells are subjected to dynamic mechanical environments which impart forces and induce cellular responses. In age-related conditions like pulmonary fibrosis, there is both an increase in tissue stiffness and an accumulation of senescent cells. While senescent cells produce a senescence-associated secretory phenotype (SASP), the impact of physical stimuli on both cellular senescence and the SASP is not well understood. Here, we show that mechanical tension, modeled using cell culture substrate rigidity, influences senescent cell markers like SA-β-gal and secretory phenotypes. Comparing human primary pulmonary fibroblasts (IMR-90) cultured on physiological (2 kPa), fibrotic (50 kPa), and plastic (approximately 3 GPa) substrates, followed by senescence induction using doxorubicin, we identified unique high-stiffness-driven secretory protein profiles using mass spectrometry and transcriptomic signatures, both showing an enrichment in collagen proteins. Consistently, clusters of p21+ cells are seen in fibrotic regions of bleomycin induced pulmonary fibrosis in mice. Computational meta-analysis of single-cell RNA sequencing datasets from human interstitial lung disease confirmed these stiffness SASP genes are highly expressed in disease fibroblasts and strongly correlate with mechanotransduction and senescence-related pathways. Thus, mechanical forces shape cell senescence and their secretory phenotypes.
Collapse
Affiliation(s)
- Huixun Du
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Jacob P Rose
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Li Guo
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | | | - Fei Wu
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Nathan Basisty
- Longitudinal Studies Section, Translational Gerontology Branch, NIA, NIH, Baltimore, Maryland, USA
| | | | | | - Nan Chen
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
| | - Veronica Chang
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
| | - Shawn Winer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, CA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Andras Nagy
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Birgit Schilling
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Daniel A Winer
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
16
|
Fan Y, Chiu A, Zhao F, George JT. Understanding the interplay between extracellular matrix topology and tumor-immune interactions: Challenges and opportunities. Oncotarget 2024; 15:768-781. [PMID: 39513932 PMCID: PMC11546212 DOI: 10.18632/oncotarget.28666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Modern cancer management comprises a variety of treatment strategies. Immunotherapy, while successful at treating many cancer subtypes, is often hindered by tumor immune evasion and T cell exhaustion as a result of an immunosuppressive tumor microenvironment (TME). In solid malignancies, the extracellular matrix (ECM) embedded within the TME plays a central role in T cell recognition and cancer growth by providing structural support and regulating cell behavior. Relative to healthy tissues, tumor associated ECM signatures include increased fiber density and alignment. These and other differentiating features contributed to variation in clinically observed tumor-specific ECM configurations, collectively referred to as Tumor-Associated Collagen Signatures (TACS) 1-3. TACS is associated with disease progression and immune evasion. This review explores our current understanding of how ECM geometry influences the behaviors of both immune cells and tumor cells, which in turn impacts treatment efficacy and cancer evolutionary progression. We discuss the effects of ECM remodeling on cancer cells and T cell behavior and review recent in silico models of cancer-immune interactions.
Collapse
Affiliation(s)
- Yijia Fan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
- Translational Medical Sciences, Texas A&M University Health Science Center, Houston, TX 77030, USA
| | - Alvis Chiu
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Jason T. George
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
- Translational Medical Sciences, Texas A&M University Health Science Center, Houston, TX 77030, USA
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA
- Department of Hematopoietic Biology and Malignancy, MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
17
|
Linke JA, Munn LL, Jain RK. Compressive stresses in cancer: characterization and implications for tumour progression and treatment. Nat Rev Cancer 2024; 24:768-791. [PMID: 39390249 DOI: 10.1038/s41568-024-00745-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: 08/20/2024] [Indexed: 10/12/2024]
Abstract
Beyond their many well-established biological aberrations, solid tumours create an abnormal physical microenvironment that fuels cancer progression and confers treatment resistance. Mechanical forces impact tumours across a range of biological sizes and timescales, from rapid events at the molecular level involved in their sensing and transmission, to slower and larger-scale events, including clonal selection, epigenetic changes, cell invasion, metastasis and immune response. Owing to challenges with studying these dynamic stimuli in biological systems, the mechanistic understanding of the effects and pathways triggered by abnormally elevated mechanical forces remains elusive, despite clear correlations with cancer pathophysiology, aggressiveness and therapeutic resistance. In this Review, we examine the emerging and diverse roles of physical forces in solid tumours and provide a comprehensive framework for understanding solid stress mechanobiology. We first review the physiological importance of mechanical forces, especially compressive stresses, and discuss their defining characteristics, biological context and relative magnitudes. We then explain how abnormal compressive stresses emerge in tumours and describe the experimental challenges in investigating these mechanically induced processes. Finally, we discuss the clinical translation of mechanotherapeutics that alleviate solid stresses and their potential to synergize with chemotherapy, radiotherapy and immunotherapies.
Collapse
Affiliation(s)
- Julia A Linke
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Wu B, Zhang B, Li B, Wu H, Jiang M. Cold and hot tumors: from molecular mechanisms to targeted therapy. Signal Transduct Target Ther 2024; 9:274. [PMID: 39420203 PMCID: PMC11491057 DOI: 10.1038/s41392-024-01979-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/20/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Immunotherapy has made significant strides in cancer treatment, particularly through immune checkpoint blockade (ICB), which has shown notable clinical benefits across various tumor types. Despite the transformative impact of ICB treatment in cancer therapy, only a minority of patients exhibit a positive response to it. In patients with solid tumors, those who respond well to ICB treatment typically demonstrate an active immune profile referred to as the "hot" (immune-inflamed) phenotype. On the other hand, non-responsive patients may exhibit a distinct "cold" (immune-desert) phenotype, differing from the features of "hot" tumors. Additionally, there is a more nuanced "excluded" immune phenotype, positioned between the "cold" and "hot" categories, known as the immune "excluded" type. Effective differentiation between "cold" and "hot" tumors, and understanding tumor intrinsic factors, immune characteristics, TME, and external factors are critical for predicting tumor response and treatment results. It is widely accepted that ICB therapy exerts a more profound effect on "hot" tumors, with limited efficacy against "cold" or "altered" tumors, necessitating combinations with other therapeutic modalities to enhance immune cell infiltration into tumor tissue and convert "cold" or "altered" tumors into "hot" ones. Therefore, aligning with the traits of "cold" and "hot" tumors, this review systematically delineates the respective immune characteristics, influencing factors, and extensively discusses varied treatment approaches and drug targets based on "cold" and "hot" tumors to assess clinical efficacy.
Collapse
Affiliation(s)
- Bo Wu
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bo Zhang
- Department of Youth League Committee, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bowen Li
- Department of Pancreatic and Gastrointestinal Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Haoqi Wu
- Department of Gynaecology and Obstetrics, The Second Hospital of Dalian Medical University, Dalian, China
| | - Meixi Jiang
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
19
|
Luo Z, Mei J, Wang X, Wang R, He Z, Geffen Y, Sun X, Zhang X, Xu J, Wan R, Feng X, Jiao C, Su X, Sun J, Chen S, Chen J, Mao W, Yang Y, Sun Y. Voluntary exercise sensitizes cancer immunotherapy via the collagen inhibition-orchestrated inflammatory tumor immune microenvironment. Cell Rep 2024; 43:114697. [PMID: 39217611 DOI: 10.1016/j.celrep.2024.114697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 07/15/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Physical activity reduces cancer-associated mortality through multiple mechanisms, including tumor immune microenvironment (TIME) reprogramming. However, whether and how physiological interventions promote anti-tumor immunity remain elusive. Here, we report that clinically relevant voluntary exercise promotes muscle-derived extracellular vesicle (EV)-associated miR-29a-3p for tumor extracellular matrix (ECM) inhibition in patients and mouse models, thereby permitting immune cell infiltration and immunotherapy. Mechanistically, an unbiased screening identifies EV-associated miR-29a-3p in response to leisure-time physical activity or voluntary exercise. MiR-29a-3p-containing EVs accumulate in tumors and downregulate collagen composition by targeting COL1A1. Gain- and loss-of-function experiments and cytometry by time of flight (CyTOF) demonstrate that myocyte-secreted miR-29a-3p promotes anti-tumor immunity. Combining immunotherapy with voluntary exercise or miR-29a-3p further enhances anti-tumor efficacy. Clinically, miR-29a-3p correlates with reduced ECM, increased T cell infiltration, and response to immunotherapy. Our work reveals the predictive value of miR-29a-3p for immunotherapy, provides mechanistic insights into exercise-induced anti-cancer immunity, and highlights the potential of voluntary exercise in sensitizing immunotherapy.
Collapse
Affiliation(s)
- Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jie Mei
- The First Clinical Medicine College, Nanjing Medical University, Nanjing 211166, China; Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China.
| | - Xianwen Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, Anhui, China
| | - Ruixin Wang
- Department of Cardiothoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Zhao He
- Department of Cardiothoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Yifat Geffen
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xiaomeng Sun
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xingyu Zhang
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Junying Xu
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Renwen Wan
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xinting Feng
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chunmeng Jiao
- Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaoping Su
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Junming Sun
- Laboratory Animal Center, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Wenjun Mao
- Department of Cardiothoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China.
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Yaying Sun
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| |
Collapse
|
20
|
Burgstaller A, Piernitzki N, Küchler N, Koch M, Kister T, Eichler H, Kraus T, Schwarz EC, Dustin ML, Lautenschläger F, Staufer O. Soft Synthetic Cells with Mobile Membrane Ligands for Ex Vivo Expansion of Therapy-Relevant T Cell Phenotypes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401844. [PMID: 38751204 DOI: 10.1002/smll.202401844] [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: 03/07/2024] [Revised: 04/29/2024] [Indexed: 10/01/2024]
Abstract
The expansion of T cells ex vivo is crucial for effective immunotherapy but currently limited by a lack of expansion approaches that closely mimic in vivo T cell activation. Taking inspiration from bottom-up synthetic biology, a new synthetic cell technology is introduced based on dispersed liquid-liquid phase-separated droplet-supported lipid bilayers (dsLBs) with tunable biochemical and biophysical characteristics, as artificial antigen presenting cells (aAPCs) for ex vivo T cell expansion. These findings obtained with the dsLB technology reveal three key insights: first, introducing laterally mobile stimulatory ligands on soft aAPCs promotes expansion of IL-4/IL-10 secreting regulatory CD8+ T cells, with a PD-1 negative phenotype, less prone to immune suppression. Second, it is demonstrated that lateral ligand mobility can mask differential T cell activation observed on substrates of varying stiffness. Third, dsLBs are applied to reveal a mechanosensitive component in bispecific Her2/CD3 T cell engager-mediated T cell activation. Based on these three insights, lateral ligand mobility, alongside receptor- and mechanosignaling, is proposed to be considered as a third crucial dimension for the design of ex vivo T cell expansion technologies.
Collapse
Affiliation(s)
- Anna Burgstaller
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, Campus E8 1, 66123, Saarbrücken, Germany
- Center for Biophysics, Saarland University, Campus Saarland, 66123, Saarbrücken, Germany
| | - Nils Piernitzki
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, Campus E8 1, 66123, Saarbrücken, Germany
- Center for Biophysics, Saarland University, Campus Saarland, 66123, Saarbrücken, Germany
| | - Nadja Küchler
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Building 48, 66421, Homburg, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
| | - Thomas Kister
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
| | - Hermann Eichler
- Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University and Saarland University Medical Center, Homburg, Germany
| | - Tobias Kraus
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Saarland University, Colloid and Interface Chemistry, 66123, Saarbrücken, Germany
| | - Eva C Schwarz
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Building 48, 66421, Homburg, Germany
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopedics, Rheumatology and, Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Franziska Lautenschläger
- Center for Biophysics, Saarland University, Campus Saarland, 66123, Saarbrücken, Germany
- Experimental Physics, Faculty of Natural Science and Technology, Saarland University, Campus Saarbrücken, 66123, Saarbrücken, Germany
| | - Oskar Staufer
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, Campus E8 1, 66123, Saarbrücken, Germany
- Center for Biophysics, Saarland University, Campus Saarland, 66123, Saarbrücken, Germany
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopedics, Rheumatology and, Musculoskeletal Sciences, University of Oxford, Oxford, UK
- Max Planck Bristol Centre for Minimal Biology, Cantock's Close, Bristol, BS8 1TS, UK
| |
Collapse
|
21
|
Karunasagara S, Taghizadeh A, Kim SH, Kim SJ, Kim YJ, Taghizadeh M, Kim MY, Oh KY, Lee JH, Kim HS, Hyun J, Kim HW. Tissue Mechanics and Hedgehog Signaling Crosstalk as a Key Epithelial-Stromal Interplay in Cancer Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400063. [PMID: 38976559 PMCID: PMC11425211 DOI: 10.1002/advs.202400063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/30/2024] [Indexed: 07/10/2024]
Abstract
Epithelial-stromal interplay through chemomechanical cues from cells and matrix propels cancer progression. Elevated tissue stiffness in potentially malignant tissues suggests a link between matrix stiffness and enhanced tumor growth. In this study, employing chronic oral/esophageal injury and cancer models, it is demonstrated that epithelial-stromal interplay through matrix stiffness and Hedgehog (Hh) signaling is key in compounding cancer development. Epithelial cells actively interact with fibroblasts, exchanging mechanoresponsive signals during the precancerous stage. Specifically, epithelial cells release Sonic Hh, activating fibroblasts to produce matrix proteins and remodeling enzymes, resulting in tissue stiffening. Subsequently, basal epithelial cells adjacent to the stiffened tissue become proliferative and undergo epithelial-to-mesenchymal transition, acquiring migratory and invasive properties, thereby promoting invasive tumor growth. Notably, transcriptomic programs of oncogenic GLI2, mechano-activated by actin cytoskeletal tension, govern this process, elucidating the crucial role of non-canonical GLI2 activation in orchestrating the proliferation and mesenchymal transition of epithelial cells. Furthermore, pharmacological intervention targeting tissue stiffening proves highly effective in slowing cancer progression. These findings underscore the impact of epithelial-stromal interplay through chemo-mechanical (Hh-stiffness) signaling in cancer development, and suggest that targeting tissue stiffness holds promise as a strategy to disrupt chemo-mechanical feedback, enabling effective cancer treatment.
Collapse
Affiliation(s)
- Shanika Karunasagara
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Ali Taghizadeh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Sang-Hyun Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Chemistry, College of Science & Technology, Dankook University, Cheonan, 31116, Republic of Korea
| | - So Jung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Yong-Jae Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Mohsen Taghizadeh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Moon-Young Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Kyu-Young Oh
- Department of Oral Pathology, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hye Sung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jeongeun Hyun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| |
Collapse
|
22
|
Sengupta K, Dillard P, Limozin L. Morphodynamics of T-lymphocytes: Scanning to spreading. Biophys J 2024; 123:2224-2233. [PMID: 38425041 PMCID: PMC11331044 DOI: 10.1016/j.bpj.2024.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/29/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024] Open
Abstract
Binding of the T cell receptor complex to its ligand, the subsequent molecular rearrangement, and the concomitant cell-scale shape changes represent the very first steps of adaptive immune recognition. The first minutes of the interaction of T cells and antigen presenting cells have been extensively scrutinized; yet, gaps remain in our understanding of how the biophysical properties of the environment may impact the sequence of events. In particular, many pioneering experiments were done on immobilized ligands and gave major insights into the process of T cell activation, whereas later experiments have indicated that ligand mobility was of paramount importance, especially to enable the formation of T cell receptor clusters. Systematic experiments to compare and reconcile the two schools are still lacking. Furthermore, recent investigations using compliant substrates have elucidated other intriguing aspects of T cell mechanics. Here we review experiments on interaction of T cells with planar artificial antigen presenting cells to explore the impact of mechanics on adhesion and actin morphodynamics during the spreading process. We enumerate a sequence tracing first contact to final spread state that is consistent with current understanding. Finally, we interpret the presented experimental results in light of a mechanical model that captures all the different morphodynamic states.
Collapse
Affiliation(s)
- Kheya Sengupta
- Aix-Marseille Université, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France.
| | - Pierre Dillard
- Aix-Marseille Université, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France; Aix-Marseille Université, CNRS, INSERM, LAI, Turing Centre for Living Systems, Marseille, France
| | - Laurent Limozin
- Aix-Marseille Université, CNRS, INSERM, LAI, Turing Centre for Living Systems, Marseille, France.
| |
Collapse
|
23
|
Rogers J, Bajur AT, Salaita K, Spillane KM. Mechanical control of antigen detection and discrimination by T and B cell receptors. Biophys J 2024; 123:2234-2255. [PMID: 38794795 PMCID: PMC11331051 DOI: 10.1016/j.bpj.2024.05.020] [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: 02/14/2024] [Revised: 05/10/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024] Open
Abstract
The adaptive immune response is orchestrated by just two cell types, T cells and B cells. Both cells possess the remarkable ability to recognize virtually any antigen through their respective antigen receptors-the T cell receptor (TCR) and B cell receptor (BCR). Despite extensive investigations into the biochemical signaling events triggered by antigen recognition in these cells, our ability to predict or control the outcome of T and B cell activation remains elusive. This challenge is compounded by the sensitivity of T and B cells to the biophysical properties of antigens and the cells presenting them-a phenomenon we are just beginning to understand. Recent insights underscore the central role of mechanical forces in this process, governing the conformation, signaling activity, and spatial organization of TCRs and BCRs within the cell membrane, ultimately eliciting distinct cellular responses. Traditionally, T cells and B cells have been studied independently, with researchers working in parallel to decipher the mechanisms of activation. While these investigations have unveiled many overlaps in how these cell types sense and respond to antigens, notable differences exist. To fully grasp their biology and harness it for therapeutic purposes, these distinctions must be considered. This review compares and contrasts the TCR and BCR, placing emphasis on the role of mechanical force in regulating the activity of both receptors to shape cellular and humoral adaptive immune responses.
Collapse
Affiliation(s)
- Jhordan Rogers
- Department of Chemistry, Emory University, Atlanta, Georgia
| | - Anna T Bajur
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, Georgia; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia.
| | - Katelyn M Spillane
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom; Department of Life Sciences, Imperial College London, London, United Kingdom.
| |
Collapse
|
24
|
Yin Y, Feng W, Chen J, Chen X, Wang G, Wang S, Xu X, Nie Y, Fan D, Wu K, Xia L. Immunosuppressive tumor microenvironment in the progression, metastasis, and therapy of hepatocellular carcinoma: from bench to bedside. Exp Hematol Oncol 2024; 13:72. [PMID: 39085965 PMCID: PMC11292955 DOI: 10.1186/s40164-024-00539-x] [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: 02/27/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous malignancy with high incidence, recurrence, and metastasis rates. The emergence of immunotherapy has improved the treatment of advanced HCC, but problems such as drug resistance and immune-related adverse events still exist in clinical practice. The immunosuppressive tumor microenvironment (TME) of HCC restricts the efficacy of immunotherapy and is essential for HCC progression and metastasis. Therefore, it is necessary to elucidate the mechanisms behind immunosuppressive TME to develop and apply immunotherapy. This review systematically summarizes the pathogenesis of HCC, the formation of the highly heterogeneous TME, and the mechanisms by which the immunosuppressive TME accelerates HCC progression and metastasis. We also review the status of HCC immunotherapy and further discuss the existing challenges and potential therapeutic strategies targeting immunosuppressive TME. We hope to inspire optimizing and innovating immunotherapeutic strategies by comprehensively understanding the structure and function of immunosuppressive TME in HCC.
Collapse
Affiliation(s)
- Yue Yin
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Weibo Feng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Jie Chen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Xilang Chen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Guodong Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xiao Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yongzhan Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Daiming Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Kaichun Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Limin Xia
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China.
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
25
|
Rodriguez BL, Huang J, Gibson L, Fradette JJ, Chen HIH, Koyano K, Cortez C, Li B, Ho C, Ashique AM, Lin VY, Crawley S, Roda JM, Chen P, Fan B, Kim J, Sissons J, Sitrin J, Kaplan DD, Gibbons DL, Rivera LB. Antitumor Activity of a Novel LAIR1 Antagonist in Combination with Anti-PD1 to Treat Collagen-Rich Solid Tumors. Mol Cancer Ther 2024; 23:1144-1158. [PMID: 38648067 PMCID: PMC11293989 DOI: 10.1158/1535-7163.mct-23-0866] [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: 12/06/2023] [Revised: 03/05/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024]
Abstract
We recently reported that resistance to PD-1 blockade in a refractory lung cancer-derived model involved increased collagen deposition and the collagen-binding inhibitory receptor leukocyte-associated immunoglobulin-like receptor 1 (LAIR1). Thus, we hypothesized that LAIR1 and collagen cooperated to suppress therapeutic response. In this study, we report that LAIR1 is associated with tumor stroma and is highly expressed by intratumoral myeloid cells in both human tumors and mouse models of cancer. Stroma-associated myeloid cells exhibit a suppressive phenotype and correlate with LAIR1 expression in human cancer. NGM438, a novel humanized LAIR1 antagonist mAb, elicits myeloid inflammation and allogeneic T-cell responses by binding to LAIR1 and blocking collagen engagement. Furthermore, a mouse-reactive NGM438 surrogate antibody sensitized refractory KP mouse lung tumors to anti-PD-1 therapy and resulted in increased intratumoral CD8+ T-cell content and inflammatory gene expression. These data place LAIR1 at the intersection of stroma and suppressive myeloid cells and support the notion that blockade of the LAIR1/collagen axis can potentially address resistance to checkpoint inhibitor therapy in the clinic.
Collapse
Affiliation(s)
- B. Leticia Rodriguez
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiawei Huang
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| | - Laura Gibson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jared J. Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hung-I H. Chen
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| | - Kikuye Koyano
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| | - Czrina Cortez
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| | - Betty Li
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| | - Carmence Ho
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| | | | - Vicky Y. Lin
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| | | | - Julie M. Roda
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| | - Peirong Chen
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| | - Bin Fan
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| | - Jeong Kim
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| | - James Sissons
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| | | | | | - Don L. Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lee B. Rivera
- NGM Biopharmaceuticals Inc. South San Francisco, CA, USA
| |
Collapse
|
26
|
Pathni A, Wagh K, Rey-Suarez I, Upadhyaya A. Mechanical regulation of lymphocyte activation and function. J Cell Sci 2024; 137:jcs219030. [PMID: 38995113 PMCID: PMC11267459 DOI: 10.1242/jcs.219030] [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] [Indexed: 07/13/2024] Open
Abstract
Mechanosensing, or how cells sense and respond to the physical environment, is crucial for many aspects of biological function, ranging from cell movement during development to cancer metastasis, the immune response and gene expression driving cell fate determination. Relevant physical stimuli include the stiffness of the extracellular matrix, contractile forces, shear flows in blood vessels, complex topography of the cellular microenvironment and membrane protein mobility. Although mechanosensing has been more widely studied in non-immune cells, it has become increasingly clear that physical cues profoundly affect the signaling function of cells of the immune system. In this Review, we summarize recent studies on mechanical regulation of immune cells, specifically lymphocytes, and explore how the force-generating cytoskeletal machinery might mediate mechanosensing. We discuss general principles governing mechanical regulation of lymphocyte function, spanning from the molecular scale of receptor activation to cellular responses to mechanical stimuli.
Collapse
Affiliation(s)
- Aashli Pathni
- Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742, USA
| | - Kaustubh Wagh
- Department of Physics, University of Maryland, College Park, MD 20742, USA
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ivan Rey-Suarez
- Insitute for Physical Science and Technology, University of Maryland, College Park, MD 20742, USA
- Microcore, Universidad de Los Andes, Bogota, DC 111711, USA
| | - Arpita Upadhyaya
- Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742, USA
- Department of Physics, University of Maryland, College Park, MD 20742, USA
- Insitute for Physical Science and Technology, University of Maryland, College Park, MD 20742, USA
- Biophysics Program, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
27
|
Mashayekhi K, Khazaie K, Faubion WA, Kim GB. Biomaterial-enhanced treg cell immunotherapy: A promising approach for transplant medicine and autoimmune disease treatment. Bioact Mater 2024; 37:269-298. [PMID: 38694761 PMCID: PMC11061617 DOI: 10.1016/j.bioactmat.2024.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 05/04/2024] Open
Abstract
Regulatory T cells (Tregs) are crucial for preserving tolerance in the body, rendering Treg immunotherapy a promising treatment option for both organ transplants and autoimmune diseases. Presently, organ transplant recipients must undergo lifelong immunosuppression to prevent allograft rejection, while autoimmune disorders lack definitive cures. In the last years, there has been notable advancement in comprehending the biology of both antigen-specific and polyclonal Tregs. Clinical trials involving Tregs have demonstrated their safety and effectiveness. To maximize the efficacy of Treg immunotherapy, it is essential for these cells to migrate to specific target tissues, maintain stability within local organs, bolster their suppressive capabilities, and ensure their intended function's longevity. In pursuit of these goals, the utilization of biomaterials emerges as an attractive supportive strategy for Treg immunotherapy in addressing these challenges. As a result, the prospect of employing biomaterial-enhanced Treg immunotherapy holds tremendous promise as a treatment option for organ transplant recipients and individuals grappling with autoimmune diseases in the near future. This paper introduces strategies based on biomaterial-assisted Treg immunotherapy to enhance transplant medicine and autoimmune treatments.
Collapse
Affiliation(s)
- Kazem Mashayekhi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - William A. Faubion
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, USA
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Gloria B. Kim
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
28
|
Shi L, Lim JY, Kam LC. Improving regulatory T cell production through mechanosensing. J Biomed Mater Res A 2024; 112:1138-1148. [PMID: 38450935 PMCID: PMC11065567 DOI: 10.1002/jbm.a.37702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024]
Abstract
Induced Tregs (iTregs) have great promise in adoptive immunotherapy for treatment of autoimmune diseases. This report investigates the impacts of substrate stiffness on human Treg induction, providing a powerful yet simple approach to improving production of these cells. Conventional CD4+ human T cells were activated on materials of different elastic modulus and cultured under suppressive conditions. Enhanced Treg induction was observed on softer materials as early as 3 days following activation and persisted for multiple weeks. Substrate stiffness also affected epigenetic modification of Treg specific genes and Treg suppressive capacity. Tregs induced on substrates of an optimal stiffness balance quantity and suppressive quality.
Collapse
Affiliation(s)
- Lingting Shi
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Jee Yoon Lim
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - Lance C. Kam
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| |
Collapse
|
29
|
Golo M, Newman PLH, Kempe D, Biro M. Mechanoimmunology in the solid tumor microenvironment. Biochem Soc Trans 2024; 52:1489-1502. [PMID: 38856041 DOI: 10.1042/bst20231427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
The tumor microenvironment (TME) is a complex and dynamic ecosystem that adjoins the cancer cells within solid tumors and comprises distinct components such as extracellular matrix, stromal and immune cells, blood vessels, and an abundance of signaling molecules. In recent years, the mechanical properties of the TME have emerged as critical determinants of tumor progression and therapeutic response. Aberrant mechanical cues, including altered tissue architecture and stiffness, contribute to tumor progression, metastasis, and resistance to treatment. Moreover, burgeoning immunotherapies hold great promise for harnessing the immune system to target and eliminate solid malignancies; however, their success is hindered by the hostile mechanical landscape of the TME, which can impede immune cell infiltration, function, and persistence. Consequently, understanding TME mechanoimmunology - the interplay between mechanical forces and immune cell behavior - is essential for developing effective solid cancer therapies. Here, we review the role of TME mechanics in tumor immunology, focusing on recent therapeutic interventions aimed at modulating the mechanical properties of the TME to potentiate T cell immunotherapies, and innovative assays tailored to evaluate their clinical efficacy.
Collapse
Affiliation(s)
- Matteo Golo
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Peter L H Newman
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Daryan Kempe
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Maté Biro
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
30
|
Kasputis T, Hosmer KE, He Y, Chen J. Ensuring food safety: Microfluidic-based approaches for the detection of food contaminants. ANALYTICAL SCIENCE ADVANCES 2024; 5:e2400003. [PMID: 38948318 PMCID: PMC11210746 DOI: 10.1002/ansa.202400003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 07/02/2024]
Abstract
Detecting foodborne contamination is a critical challenge in ensuring food safety and preventing human suffering and economic losses. Contaminated food, comprising biological agents (e.g. bacteria, viruses and fungi) and chemicals (e.g. toxins, allergens, antibiotics and heavy metals), poses significant risks to public health. Microfluidic technology has emerged as a transformative solution, revolutionizing the detection of contaminants with precise and efficient methodologies. By manipulating minute volumes of fluid on miniaturized systems, microfluidics enables the creation of portable chips for biosensing applications. Advancements from early glass and silicon devices to modern polymers and cellulose-based chips have significantly enhanced microfluidic technology, offering adaptability, flexibility, cost-effectiveness and biocompatibility. Microfluidic systems integrate seamlessly with various biosensing reactions, facilitating nucleic acid amplification, target analyte recognition and accurate signal readouts. As research progresses, microfluidic technology is poised to play a pivotal role in addressing evolving challenges in the detection of foodborne contaminants. In this short review, we delve into various manufacturing materials for state-of-the-art microfluidic devices, including inorganics, elastomers, thermoplastics and paper. Additionally, we examine several applications where microfluidic technology offers unique advantages in the detection of food contaminants, including bacteria, viruses, fungi, allergens and more. This review underscores the significant advancement of microfluidic technology and its pivotal role in advancing the detection and mitigation of foodborne contaminants.
Collapse
Affiliation(s)
- Tom Kasputis
- Department of Biological Systems EngineeringVirginia TechBlacksburgVirginiaUSA
| | | | - Yawen He
- Department of Biological Systems EngineeringVirginia TechBlacksburgVirginiaUSA
| | - Juhong Chen
- Department of Biological Systems EngineeringVirginia TechBlacksburgVirginiaUSA
- Department of BioengineeringUniversity of CaliforniaRiversideCaliforniaUSA
| |
Collapse
|
31
|
Benmelech S, Le T, McKay M, Nam J, Subramaniam K, Tellez D, Vlasak G, Mak M. Biophysical and biochemical aspects of immune cell-tumor microenvironment interactions. APL Bioeng 2024; 8:021502. [PMID: 38572312 PMCID: PMC10990568 DOI: 10.1063/5.0195244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
The tumor microenvironment (TME), composed of and influenced by a heterogeneous set of cancer cells and an extracellular matrix, plays a crucial role in cancer progression. The biophysical aspects of the TME (namely, its architecture and mechanics) regulate interactions and spatial distributions of cancer cells and immune cells. In this review, we discuss the factors of the TME-notably, the extracellular matrix, as well as tumor and stromal cells-that contribute to a pro-tumor, immunosuppressive response. We then discuss the ways in which cells of the innate and adaptive immune systems respond to tumors from both biochemical and biophysical perspectives, with increased focus on CD8+ and CD4+ T cells. Building upon this information, we turn to immune-based antitumor interventions-specifically, recent biophysical breakthroughs aimed at improving CAR-T cell therapy.
Collapse
Affiliation(s)
- Shoham Benmelech
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Thien Le
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Maggie McKay
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Jungmin Nam
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Krupakar Subramaniam
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06511, USA
| | - Daniela Tellez
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Grace Vlasak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| |
Collapse
|
32
|
Wang X, Fernandes SM, Brown JR, Kam LC. Assaying and classifying T cell function by cell morphology. BIOMEDINFORMATICS 2024; 4:1144-1154. [PMID: 39525274 PMCID: PMC11542667 DOI: 10.3390/biomedinformatics4020063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Immune cell function varies tremendously between individuals, posing a major challenge to emerging cellular immunotherapies. This report pursues the use of cell morphology as an indicator of high-level T cell function. Short-term spreading of T cells on planar, elastic surfaces was quantified by 11 morphological parameters and analyzed to identify effects of both intrinsic and extrinsic factors. Our findings identified morphological features that varied between T cells isolated from healthy donors and those from patients being treated for Chronic Lymphocytic Leukemia (CLL). This approach also identified differences between cell responses to substrates of different elastic modulus. Combining multiple features through a machine learning approach such as Decision Tree or Random Forest provided an effective means for identifying whether T cells came from healthy or CLL donors. Further development of this approach could lead to a rapid assay of T cell function to guide cellular immunotherapy.
Collapse
Affiliation(s)
- Xin Wang
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Stacey M. Fernandes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Jennifer R. Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Lance C. Kam
- Department of Biomedical Engineering, Columbia University, New York, NY
| |
Collapse
|
33
|
Zeng Q, Xu B, Deng J, Shang K, Guo Z, Wu S. Optimization of polydimethylsiloxane (PDMS) surface chemical modification and formulation for improved T cell activation and expansion. Colloids Surf B Biointerfaces 2024; 239:113977. [PMID: 38776594 DOI: 10.1016/j.colsurfb.2024.113977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/30/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024]
Abstract
Adoptive T cell therapy has undergone remarkable advancements in recent decades; nevertheless, the rapid and effective ex vivo expansion of tumor-reactive T cells remains a formidable challenge, limiting their clinical application. Artificial antigen-presenting substrates represent a promising avenue for enhancing the efficiency of adoptive immunotherapy and fostering T cell expansion. These substrates offer significant potential by providing flexibility and modularity in the design of tailored stimulatory environments. Polydimethylsiloxane (PDMS) silicone elastomer stands as a widely utilized biomaterial for exploring the varying sensitivity of T cell activation to substrate properties. This paper explores the optimization of PDMS surface modification and formulation to create customized stimulatory surfaces with the goal of enhancing T cell expansion. By employing soft PDMS elastomer functionalized through silanization and activating agent, coupled with site-directed protein immobilization techniques, a novel T cell stimulatory platform is introduced, facilitating T cell activation and proliferation. Notably, our findings underscore that softer modified elastomers (Young' modulus E∼300 kPa) exhibit superior efficacy in stimulating and activating mouse CD4+ T cells compared to their stiffer counterparts (E∼3 MPa). Furthermore, softened modified PDMS substrates demonstrate enhanced capabilities in T cell expansion and Th1 differentiation, offering promising insights for the advancement of T cell-based immunotherapy.
Collapse
Affiliation(s)
- Qiongjiao Zeng
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Bowen Xu
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China; Department of Clinical Laboratory, Third Affiliated Hospital of Naval Medical University, Shanghai 200438, China
| | - Jiewen Deng
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Kun Shang
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Zhenhong Guo
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China.
| | - Shuqing Wu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
34
|
Valentić B, Kelly A, Shestov AA, Gan Z, Shen F, Chatoff A, Jaccard A, Crispim CV, Scholler J, Heeke S, Snyder NW, Ghassemi S, Jones N, Gill S, O'Connor RS. The Glucose Transporter 5 Enhances CAR-T Cell Metabolic Function and Anti-tumour Durability. RESEARCH SQUARE 2024:rs.3.rs-4342820. [PMID: 38766088 PMCID: PMC11100898 DOI: 10.21203/rs.3.rs-4342820/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Activated T cells undergo a metabolic shift to aerobic glycolysis to support the energetic demands of proliferation, differentiation, and cytolytic function. Transmembrane glucose flux is facilitated by glucose transporters (GLUT) that play a vital role in T cell metabolic reprogramming and anti-tumour function. GLUT isoforms are regulated at the level of expression and subcellular distribution. GLUTs also display preferential selectivity for carbohydrate macronutrients including glucose, galactose, and fructose. GLUT5, which selectively transports fructose over glucose, has never been explored as a genetic engineering strategy to enhance CAR-T cells in fructose-rich tumour environments. Fructose levels are significantly elevated in the bone marrow and the plasma of acute myeloid leukaemia (AML) patients. Here, we demonstrate that the expression of wild-type GLUT5 restores T cell metabolic fitness in glucose-free, high fructose conditions. We find that fructose supports maximal glycolytic capacity and ATP replenishment rates in GLUT5-expressing T cells. Using steady state tracer technology, we show that 13C6 fructose supports glycolytic reprogramming and TCA anaplerosis in CAR-T cells undergoing log phase expansion. In cytotoxicity assays, GLUT5 rescues T cell cytolytic function in glucose-free medium. The fructose/GLUT5 metabolic axis also supports maximal migratory velocity, which provides mechanistic insight into why GLUT5-expressing CAR-Ts have superior effector function as they undergo "hit-and-run" serial killing. These findings translate to superior anti-tumour function in a xenograft model of AML. In fact, we found that GLUT5 enhances CAR-T cell anti-tumour function in vivo without any need for fructose intervention. Accordingly, we hypothesize that GLUT5 is sufficient to enhance CAR-T resilience by increasing the cells' competitiveness for glucose at physiologic metabolite levels. Our findings have immediate translational relevance by providing the first evidence that GLUT5 confers a competitive edge in a fructose-enriched milieu, and is a novel approach to overcome glucose depletion in hostile tumour microenvironments (TMEs).
Collapse
Affiliation(s)
- Bakir Valentić
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andre Kelly
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexander A Shestov
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhiyang Gan
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Feng Shen
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Haematology-Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Adam Chatoff
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Alison Jaccard
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Claudia V Crispim
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - John Scholler
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Simon Heeke
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nathaniel W Snyder
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Saba Ghassemi
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas Jones
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK
| | - Saar Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Haematology-Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roddy S O'Connor
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
35
|
Mei J, Cai Y, Xu R, Li Q, Chu J, Luo Z, Sun Y, Shi Y, Xu J, Li D, Liang S, Jiang Y, Liu J, Qian Z, Zhou J, Wan M, Yang Y, Zhu Y, Zhang Y, Yin Y. Conserved immuno-collagenic subtypes predict response to immune checkpoint blockade. Cancer Commun (Lond) 2024; 44:554-575. [PMID: 38507505 PMCID: PMC11110954 DOI: 10.1002/cac2.12538] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/06/2024] [Accepted: 03/10/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) has revolutionized the treatment of various cancer types. Despite significant preclinical advancements in understanding mechanisms, identifying the molecular basis and predictive biomarkers for clinical ICB responses remains challenging. Recent evidence, both preclinical and clinical, underscores the pivotal role of the extracellular matrix (ECM) in modulating immune cell infiltration and behaviors. This study aimed to create an innovative classifier that leverages ECM characteristics to enhance the effectiveness of ICB therapy. METHODS We analyzed transcriptomic collagen activity and immune signatures in 649 patients with cancer undergoing ICB therapy. This analysis led to the identification of three distinct immuno-collagenic subtypes predictive of ICB responses. We validated these subtypes using the transcriptome data from 9,363 cancer patients from The Cancer Genome Atlas (TCGA) dataset and 1,084 in-house samples. Additionally, novel therapeutic targets were identified based on these established immuno-collagenic subtypes. RESULTS Our categorization divided tumors into three subtypes: "soft & hot" (low collagen activity and high immune infiltration), "armored & cold" (high collagen activity and low immune infiltration), and "quiescent" (low collagen activity and immune infiltration). Notably, "soft & hot" tumors exhibited the most robust response to ICB therapy across various cancer types. Mechanistically, inhibiting collagen augmented the response to ICB in preclinical models. Furthermore, these subtypes demonstrated associations with immune activity and prognostic predictive potential across multiple cancer types. Additionally, an unbiased approach identified B7 homolog 3 (B7-H3), an available drug target, as strongly expressed in "armored & cold" tumors, relating with poor prognosis. CONCLUSION This study introduces histopathology-based universal immuno-collagenic subtypes capable of predicting ICB responses across diverse cancer types. These findings offer insights that could contribute to tailoring personalized immunotherapeutic strategies for patients with cancer.
Collapse
Affiliation(s)
- Jie Mei
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsuP. R. China
| | - Yun Cai
- Departments of GynecologyWuxi Maternal and Child Health Care Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuP. R. China
| | - Rui Xu
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsuP. R. China
| | - Qing Li
- Departments of OncologyXuzhou Central HospitalThe Xuzhou School of Clinical Medicine of Nanjing Medical UniversityXuzhouJiangsuP. R. China
| | - Jiahui Chu
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsuP. R. China
| | - Zhiwen Luo
- Department of Sports MedicineHuashan Hospital Affiliated to Fudan UniversityShanghaiP. R. China
| | - Yaying Sun
- Department of Sports MedicineShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Yuxin Shi
- Departments of OncologyThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuP. R. China
| | - Junying Xu
- Departments of OncologyThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuP. R. China
| | - Di Li
- Shanghai Outdo Biotech Co., Ltd., National Engineering Center for BiochipShanghaiP. R. China
| | - Shuai Liang
- Departments of OncologyThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuP. R. China
| | - Ying Jiang
- Departments of GynecologyWuxi Maternity and Child Health Care HospitalAffiliated Women's Hospital of Jiangnan UniversityWuxiJiangsuP. R. China
| | - Jiayu Liu
- Departments of GynecologyWuxi Maternity and Child Health Care HospitalAffiliated Women's Hospital of Jiangnan UniversityWuxiJiangsuP. R. China
| | - Zhiwen Qian
- Departments of GynecologyWuxi Maternal and Child Health Care Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuP. R. China
| | - Jiaofeng Zhou
- Department of PhysiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjingJiangsuP. R. China
| | - Mengyun Wan
- Department of PhysiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjingJiangsuP. R. China
| | - Yunlong Yang
- Department of Cellular and Genetic MedicineSchool of Basic Medical Sciences, Fudan UniversityShanghaiP. R. China
| | - Yichao Zhu
- Department of PhysiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjingJiangsuP. R. China
| | - Yan Zhang
- Departments of GynecologyWuxi Maternal and Child Health Care Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuP. R. China
- Departments of GynecologyWuxi Maternity and Child Health Care HospitalAffiliated Women's Hospital of Jiangnan UniversityWuxiJiangsuP. R. China
| | - Yongmei Yin
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical UniversityNanjingJiangsuP. R. China
| |
Collapse
|
36
|
Mai Z, Lin Y, Lin P, Zhao X, Cui L. Modulating extracellular matrix stiffness: a strategic approach to boost cancer immunotherapy. Cell Death Dis 2024; 15:307. [PMID: 38693104 PMCID: PMC11063215 DOI: 10.1038/s41419-024-06697-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024]
Abstract
The interplay between extracellular matrix (ECM) stiffness and the tumor microenvironment is increasingly recognized as a critical factor in cancer progression and the efficacy of immunotherapy. This review comprehensively discusses the key factors regulating ECM remodeling, including the activation of cancer-associated fibroblasts and the accumulation and crosslinking of ECM proteins. Furthermore, it provides a detailed exploration of how ECM stiffness influences the behaviors of both tumor and immune cells. Significantly, the impact of ECM stiffness on the response to various immunotherapy strategies, such as immune checkpoint blockade, adoptive cell therapy, oncolytic virus therapy, and therapeutic cancer vaccines, is thoroughly examined. The review also addresses the challenges in translating research findings into clinical practice, highlighting the need for more precise biomaterials that accurately mimic the ECM and the development of novel therapeutic strategies. The insights offered aim to guide future research, with the potential to enhance the effectiveness of cancer immunotherapy modalities.
Collapse
Affiliation(s)
- Zizhao Mai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
37
|
Zeng Q, Xu B, Qian C, Li N, Guo Z, Wu S. Surface chemical modification of poly(dimethylsiloxane) for stabilizing antibody immobilization and T cell cultures. Biomater Sci 2024; 12:2369-2380. [PMID: 38498344 DOI: 10.1039/d3bm01729j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Advances in cell immunotherapy underscore the need for effective methods to produce large populations of effector T cells, driving growing interest in T-cell bioprocessing and immunoengineering. Research suggests that T cells demonstrate enhanced expansion and differentiation on soft matrices in contrast to rigid ones. Nevertheless, the influence of antibody conjugation chemistry on these processes remains largely unexplored. In this study, we examined the effect of antibody conjugation chemistry on T cell activation, expansion and differentiation using a soft and biocompatible polydimethylsiloxane (PDMS) platform. We rigorously evaluated three distinct immobilization methods, beginning with the use of amino-silane (PDMS-NH2-Ab), followed by glutaraldehyde (PDMS-CHO-Ab) or succinic acid anhydride (PDMS-COOH-Ab) activation, in addition to the conventional physical adsorption (PDMS-Ab). By employing both stable amide bonds and reducible Schiff bases, antibody conjugation significantly enhanced antibody loading and density compared to physical adsorption. Furthermore, we discovered that the PDMS-COOH-Ab surface significantly promoted IL-2 secretion, CD69 expression, and T cell expansion compared to the other groups. Moreover, we observed that both PDMS-COOH-Ab and PDMS-NH2-Ab surfaces exhibited a tendency to induce the differentiation of naïve CD4+ T cells into Th1 cells, whereas the PDMS-Ab surface elicited a Th2-biased immunological response. These findings highlight the importance of antibody conjugation chemistry in the design and development of T cell culture biomaterials. They also indicate that PDMS holds promise as a material for constructing culture platforms to modulate T cell activation, proliferation, and differentiation.
Collapse
MESH Headings
- Dimethylpolysiloxanes/chemistry
- T-Lymphocytes/immunology
- Surface Properties
- Antibodies, Immobilized/chemistry
- Antibodies, Immobilized/immunology
- Cell Differentiation/drug effects
- Animals
- Lymphocyte Activation/drug effects
- Cell Proliferation/drug effects
- Interleukin-2/metabolism
- Interleukin-2/chemistry
- Mice
- Cells, Cultured
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/chemistry
- Adsorption
- Succinic Anhydrides
Collapse
Affiliation(s)
- Qiongjiao Zeng
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Bowen Xu
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai, 200433, China.
| | - Cheng Qian
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai, 200433, China.
| | - Nan Li
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai, 200433, China.
| | - Zhenhong Guo
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai, 200433, China.
| | - Shuqing Wu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| |
Collapse
|
38
|
Lizana-Vasquez GD, Mendez-Vega J, Cappabianca D, Saha K, Torres-Lugo M. In vitro encapsulation and expansion of T and CAR-T cells using 3D synthetic thermo-responsive matrices. RSC Adv 2024; 14:13734-13747. [PMID: 38681842 PMCID: PMC11046447 DOI: 10.1039/d4ra01968g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
Suspension cell culture and rigid commercial substrates are the most common methods to clinically manufacture therapeutic CAR-T cells ex vivo. However, suspension culture and nano/micro-scale commercial substrates poorly mimic the microenvironment where T cells naturally develop, leading to profound impacts on cell proliferation and phenotype. To overcome this major challenge, macro-scale substrates can be used to emulate that environment with higher precision. This work employed a biocompatible thermo-responsive material with tailored mechanical properties as a potential synthetic macro-scale scaffold to support T cell encapsulation and culture. Cell viability, expansion, and phenotype changes were assessed to study the effect of two thermo-responsive hydrogel materials with stiffnesses of 0.5 and 17 kPa. Encapsulated Pan-T and CAR-T cells were able to grow and physically behave similar to the suspension control. Furthermore, matrix stiffness influenced T cell behavior. In the softer polymer, T cells had higher activation, differentiation, and maturation after encapsulation obtaining significant cell numbers. Even when terpolymer encapsulation affected the CAR-T cell viability and expansion, CAR T cells expressed favorable phenotypical profiles, which was supported with cytokines and lactate production. These results confirmed the biocompatibility of the thermo-responsive hydrogels and their feasibility as a promising 3D macro-scale scaffold for in vitro T cell expansion that could potentially be used for cell manufacturing process.
Collapse
Affiliation(s)
- Gaby D Lizana-Vasquez
- Deparment of Chemical Engineering, University of Puerto Rico-Mayagüez Road 108 Km. 1.0 Bo. Miradero. P.O. Box 9046 Mayagüez 00681-9046 Puerto Rico USA +1 787 832 4040 Ext. 2585
| | - Janet Mendez-Vega
- Deparment of Chemical Engineering, University of Puerto Rico-Mayagüez Road 108 Km. 1.0 Bo. Miradero. P.O. Box 9046 Mayagüez 00681-9046 Puerto Rico USA +1 787 832 4040 Ext. 2585
| | - Dan Cappabianca
- Department of Biomedical Engineering, University of Wisconsin-Madison Madison Wisconsin USA
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin-Madison Madison Wisconsin USA
| | - Madeline Torres-Lugo
- Deparment of Chemical Engineering, University of Puerto Rico-Mayagüez Road 108 Km. 1.0 Bo. Miradero. P.O. Box 9046 Mayagüez 00681-9046 Puerto Rico USA +1 787 832 4040 Ext. 2585
| |
Collapse
|
39
|
López Ruiz A, Slaughter ED, Kloxin AM, Fromen CA. Bridging the gender gap in autoimmunity with T-cell-targeted biomaterials. Curr Opin Biotechnol 2024; 86:103075. [PMID: 38377884 PMCID: PMC11578274 DOI: 10.1016/j.copbio.2024.103075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/22/2024]
Abstract
Autoimmune diseases are caused by malfunctions of the immune system and generally impact women at twice the frequency of men. Many of the most serious autoimmune diseases are accompanied by a dysregulation of T-cell phenotype, both regarding the ratio of CD4+ to CD8+ T-cells and proinflammatory versus regulatory phenotypes. Biomaterials, in the form of particles and hydrogels, have shown promise in ameliorating this dysregulation both in vivo and ex vivo. In this review, we explore the role of T-cells in autoimmune diseases, particularly those with high incidence rates in women, and evaluate the promise and efficacy of innovative biomaterial-based approaches for targeting T-cells.
Collapse
Affiliation(s)
- Aida López Ruiz
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
| | - Eric D Slaughter
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
| | - April M Kloxin
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States; Material Science and Engineering, University of Delaware, Newark, DE, United States.
| | - Catherine A Fromen
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States.
| |
Collapse
|
40
|
Mittelheisser V, Gensbittel V, Bonati L, Li W, Tang L, Goetz JG. Evidence and therapeutic implications of biomechanically regulated immunosurveillance in cancer and other diseases. NATURE NANOTECHNOLOGY 2024; 19:281-297. [PMID: 38286876 DOI: 10.1038/s41565-023-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 09/26/2023] [Indexed: 01/31/2024]
Abstract
Disease progression is usually accompanied by changes in the biochemical composition of cells and tissues and their biophysical properties. For instance, hallmarks of cancer include the stiffening of tissues caused by extracellular matrix remodelling and the softening of individual cancer cells. In this context, accumulating evidence has shown that immune cells sense and respond to mechanical signals from the environment. However, the mechanisms regulating these mechanical aspects of immune surveillance remain partially understood. The growing appreciation for the 'mechano-immunology' field has urged researchers to investigate how immune cells sense and respond to mechanical cues in various disease settings, paving the way for the development of novel engineering strategies that aim at mechanically modulating and potentiating immune cells for enhanced immunotherapies. Recent pioneer developments in this direction have laid the foundations for leveraging 'mechanical immunoengineering' strategies to treat various diseases. This Review first outlines the mechanical changes occurring during pathological progression in several diseases, including cancer, fibrosis and infection. We next highlight the mechanosensitive nature of immune cells and how mechanical forces govern the immune responses in different diseases. Finally, we discuss how targeting the biomechanical features of the disease milieu and immune cells is a promising strategy for manipulating therapeutic outcomes.
Collapse
Affiliation(s)
- Vincent Mittelheisser
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Valentin Gensbittel
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Lucia Bonati
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Weilin Li
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Materials Science and Engineering, EPFL, Lausanne, Switzerland.
| | - Jacky G Goetz
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
| |
Collapse
|
41
|
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
|
42
|
Zhu Z, Wang Q, Zeng X, Zhu S, Chen J. Validation and identification of anoikis-related lncRNA signatures for improving prognosis in clear cell renal cell carcinoma. Aging (Albany NY) 2024; 16:3915-3933. [PMID: 38385949 PMCID: PMC10929799 DOI: 10.18632/aging.205568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/21/2023] [Indexed: 02/23/2024]
Abstract
BACKGROUND Clear cell carcinoma (ccRCC) usually has a high metastasis rate and high mortality rate. To enable precise risk stratification, there is a need for novel biomarkers. As one form of apoptosis, anoikis results from the disruption of cell-cell connection or cell-ECM attachment. However, the impact of anoikis-related lncRNAs on ccRCC has not yet received adequate attention. METHODS The study utilized univariate Cox regression analysis in order to identify the overall survival (OS) associated anoikis-related lncRNAs (ARLs), followed by the LASSO algorithm for selection. On this basis, a risk model was subsequently established using five anoikis-related lncRNAs. To dig the inner molecular mechanism, KEGG, GO, and GSVA analyses were conducted. Additionally, the immune infiltration landscape was estimated using the ESTIMATE, CIBERSORT, and ssGSEA algorithms. RESULTS The study constructed a novel risk model based on five ARLs (AC092611.2, AC027601.2, AC103809.1, AL133215.2, and AL162586.1). Patients categorized as low-risk exhibited significantly better OS. Notably, the study observed marked different immune infiltration landscapes and drug sensitivity by risk stratification. Additionally, the study preliminarily explored potential signal pathways associated with risk stratification. CONCLUSION The study exhibited the crucial role of ARLs in the carcinogenesis of ccRCC, potentially through differential immune infiltration. Furthermore, the established risk model could serve as a valuable stratification factor for predicting OS prognosis.
Collapse
Affiliation(s)
- Zhenjie Zhu
- Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Qibo Wang
- Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiaowei Zeng
- Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Shaoxing Zhu
- Fujian Medical University Union Hospital, Fuzhou, China
| | - Jinchao Chen
- Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
43
|
Du W, Xia X, Hu F, Yu J. Extracellular matrix remodeling in the tumor immunity. Front Immunol 2024; 14:1340634. [PMID: 38332915 PMCID: PMC10850336 DOI: 10.3389/fimmu.2023.1340634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/28/2023] [Indexed: 02/10/2024] Open
Abstract
The extracellular matrix (ECM) is a significant constituent of tumors, fulfilling various essential functions such as providing mechanical support, influencing the microenvironment, and serving as a reservoir for signaling molecules. The abundance and degree of cross-linking of ECM components are critical determinants of tissue stiffness. In the process of tumorigenesis, the interaction between ECM and immune cells within the tumor microenvironment (TME) frequently leads to ECM stiffness, thereby disrupting normal mechanotransduction and promoting malignant progression. Therefore, acquiring a thorough comprehension of the dysregulation of ECM within the TME would significantly aid in the identification of potential therapeutic targets for cancer treatment. In this regard, we have compiled a comprehensive summary encompassing the following aspects: (1) the principal components of ECM and their roles in malignant conditions; (2) the intricate interaction between ECM and immune cells within the TME; and (3) the pivotal regulators governing the onco-immune response in ECM.
Collapse
Affiliation(s)
- Wei Du
- Department of Targeting Therapy and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Xueming Xia
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Fan Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jiayun Yu
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
44
|
Yan B, Liao P, Shi L, Lei P. Pan-cancer analyses of senescence-related genes in extracellular matrix characterization in cancer. Discov Oncol 2023; 14:208. [PMID: 37985530 PMCID: PMC10660488 DOI: 10.1007/s12672-023-00828-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023] Open
Abstract
PURPOSE The aged microenvironment plays a crucial role in tumor onset and progression. However, it remains unclear whether and how the aging of the extracellular matrix (ECM) influences cancer onset and progression. Furthermore, the mechanisms and implications of extracellular matrix senescence-related genes (ECM-SRGs) in pan-cancer have not been investigated. METHODS We collected profiling data from over 10,000 individuals, covering 33 cancer types, 750 small molecule drugs, and 24 immune cell types, for a thorough and systematic analysis of ECM-SRGs in cancer. RESULTS We observed a significant correlation between immune cell infiltrates and Gene Set Variation Analysis enrichment scores of ECM-SRGs in 33 cancer types. Moreover, our results revealed significant differences in immune cell infiltration among patients with copy number variations (CNV) and single nucleotide variations (SNV) in ECM-SRGs across various malignancies. Aberrant hypomethylation led to increased ECM-SRGs expression, and in specific malignancies, a connection between ECM-SRGs hypomethylation and adverse patient survival was established. The frequency of CNV and SNV in ECM-SRGs was elevated. We observed a positive correlation between CNV, SNV, and ECM-SRGs expression. Furthermore, a correlation was found between the high frequency of CNV and SNV in ECM-SRGs and poor patient survival in several cancer types. Additionally, the results demonstrated that ECM-SRGs expression could serve as a predictor of patient survival in diverse cancers. Pathway analysis unveiled the role of ECM-SRGs in activating EMT, apoptosis, and the RAS/MAPK signaling pathway while suppressing the cell cycle, hormone AR, and the response to DNA damage signaling pathway. Finally, we conducted searches in the "Genomics of Drug Sensitivity in Cancer" and "Genomics of Therapeutics Response Portal" databases, identifying several drugs that target ECM-SRGs. CONCLUSIONS We conducted a comprehensive evaluation of the genomes and immunogenomics of ECM-SRGs, along with their clinical features in 33 solid tumors. This may provide insights into the relationship between ECM-SRGs and tumorigenesis. Consequently, targeting these ECM-SRGs holds promise as a clinical approach for cancer treatment.
Collapse
Affiliation(s)
- Bo Yan
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Pan Liao
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- The School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Liqiu Shi
- Inner Mongolia Forestry General Hospital, 81 Lincheng North Road, Yakeshi, 022150, Inner Mongolia, China
| | - Ping Lei
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
- The School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
45
|
Adu-Berchie K, Liu Y, Zhang DKY, Freedman BR, Brockman JM, Vining KH, Nerger BA, Garmilla A, Mooney DJ. Generation of functionally distinct T-cell populations by altering the viscoelasticity of their extracellular matrix. Nat Biomed Eng 2023; 7:1374-1391. [PMID: 37365267 PMCID: PMC10749992 DOI: 10.1038/s41551-023-01052-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/05/2023] [Indexed: 06/28/2023]
Abstract
The efficacy of adoptive T-cell therapies largely depends on the generation of T-cell populations that provide rapid effector function and long-term protective immunity. Yet it is becoming clearer that the phenotypes and functions of T cells are inherently linked to their localization in tissues. Here we show that functionally distinct T-cell populations can be generated from T cells that received the same stimulation by altering the viscoelasticity of their surrounding extracellular matrix (ECM). By using a model ECM based on a norbornene-modified collagen type I whose viscoelasticity can be adjusted independently from its bulk stiffness by varying the degree of covalent crosslinking via a bioorthogonal click reaction with tetrazine moieties, we show that ECM viscoelasticity regulates T-cell phenotype and function via the activator-protein-1 signalling pathway, a critical regulator of T-cell activation and fate. Our observations are consistent with the tissue-dependent gene-expression profiles of T cells isolated from mechanically distinct tissues from patients with cancer or fibrosis, and suggest that matrix viscoelasticity could be leveraged when generating T-cell products for therapeutic applications.
Collapse
Affiliation(s)
- Kwasi Adu-Berchie
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Yutong Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - David K Y Zhang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Benjamin R Freedman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Joshua M Brockman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Kyle H Vining
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Preventative and Restorative Sciences, School of Dental Medicine, and Department of Materials Science and Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Bryan A Nerger
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | | | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
46
|
Manca F, Eich G, N'Dao O, Normand L, Sengupta K, Limozin L, Puech PH. Probing mechanical interaction of immune receptors and cytoskeleton by membrane nanotube extraction. Sci Rep 2023; 13:15652. [PMID: 37730849 PMCID: PMC10511455 DOI: 10.1038/s41598-023-42599-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/12/2023] [Indexed: 09/22/2023] Open
Abstract
The role of force application in immune cell recognition is now well established, the force being transmitted between the actin cytoskeleton to the anchoring ligands through receptors such as integrins. In this chain, the mechanics of the cytoskeleton to receptor link, though clearly crucial, remains poorly understood. To probe this link, we combine mechanical extraction of membrane tubes from T cells using optical tweezers, and fitting of the resulting force curves with a viscoelastic model taking into account the cell and relevant molecules. We solicit this link using four different antibodies against various membrane bound receptors: antiCD3 to target the T Cell Receptor (TCR) complex, antiCD45 for the long sugar CD45, and two clones of antiCD11 targeting open or closed conformation of LFA1 integrins. Upon disruption of the cytoskeleton, the stiffness of the link changes for two of the receptors, exposing the existence of a receptor to cytoskeleton link-namely TCR-complex and open LFA1, and does not change for the other two where a weaker link was expected. Our integrated approach allows us to probe, for the first time, the mechanics of the intracellular receptor-cytoskeleton link in immune cells.
Collapse
Affiliation(s)
- Fabio Manca
- CNRS, INSERM, Laboratoire Adhesion et Inflammation (LAI), Aix Marseille University, 13009, Marseille, France.
- CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINaM), Aix Marseille University, 13009, Marseille, France.
- Turing Center for Living Systems (CENTURI), 13009, Marseille, France.
| | - Gautier Eich
- CNRS, INSERM, Laboratoire Adhesion et Inflammation (LAI), Aix Marseille University, 13009, Marseille, France
| | - Omar N'Dao
- CNRS, INSERM, Laboratoire Adhesion et Inflammation (LAI), Aix Marseille University, 13009, Marseille, France
| | - Lucie Normand
- CNRS, INSERM, Laboratoire Adhesion et Inflammation (LAI), Aix Marseille University, 13009, Marseille, France
| | - Kheya Sengupta
- CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINaM), Aix Marseille University, 13009, Marseille, France.
- Turing Center for Living Systems (CENTURI), 13009, Marseille, France.
| | - Laurent Limozin
- CNRS, INSERM, Laboratoire Adhesion et Inflammation (LAI), Aix Marseille University, 13009, Marseille, France.
- Turing Center for Living Systems (CENTURI), 13009, Marseille, France.
| | - Pierre-Henri Puech
- CNRS, INSERM, Laboratoire Adhesion et Inflammation (LAI), Aix Marseille University, 13009, Marseille, France.
- Turing Center for Living Systems (CENTURI), 13009, Marseille, France.
| |
Collapse
|
47
|
Viúdez-Pareja C, Kreft E, García-Caballero M. Immunomodulatory properties of the lymphatic endothelium in the tumor microenvironment. Front Immunol 2023; 14:1235812. [PMID: 37744339 PMCID: PMC10512957 DOI: 10.3389/fimmu.2023.1235812] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/08/2023] [Indexed: 09/26/2023] Open
Abstract
The tumor microenvironment (TME) is an intricate complex and dynamic structure composed of various cell types, including tumor, stromal and immune cells. Within this complex network, lymphatic endothelial cells (LECs) play a crucial role in regulating immune responses and influencing tumor progression and metastatic dissemination to lymph node and distant organs. Interestingly, LECs possess unique immunomodulatory properties that can either promote or inhibit anti-tumor immune responses. In fact, tumor-associated lymphangiogenesis can facilitate tumor cell dissemination and metastasis supporting immunoevasion, but also, different molecular mechanisms involved in LEC-mediated anti-tumor immunity have been already described. In this context, the crosstalk between cancer cells, LECs and immune cells and how this communication can shape the immune landscape in the TME is gaining increased interest in recent years. In this review, we present a comprehensive and updated report about the immunomodulatory properties of the lymphatic endothelium within the TME, with special focus on primary tumors and tumor-draining lymph nodes. Furthermore, we outline emerging research investigating the potential therapeutic strategies targeting the lymphatic endothelium to enhance anti-tumor immune responses. Understanding the intricate mechanisms involved in LEC-mediated immune modulation in the TME opens up new possibilities for the development of innovative approaches to fight cancer.
Collapse
Affiliation(s)
- Cristina Viúdez-Pareja
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Andalucía Tech, University of Málaga, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| | - Ewa Kreft
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Andalucía Tech, University of Málaga, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| | - Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Andalucía Tech, University of Málaga, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| |
Collapse
|
48
|
Joseph JP, Gugulothu SB, Nandi D, Chatterjee K. Mechanical Properties Affect Primary T Cell Activation in 3D Bioprinted Hydrogels. ACS Macro Lett 2023; 12:1085-1093. [PMID: 37466277 DOI: 10.1021/acsmacrolett.3c00271] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
T cells play a critical role in the adaptive immune response of the body, especially against intracellular pathogens and cancer. In vitro, T cell activation studies typically employ planar (two-dimensional, 2D) culture systems that do not mimic native cell-to-extracellular matrix (ECM) interactions, which influence activation. The goal of this work was to study T cell responses in a cell line (EL4) and primary mouse T cells in three-dimensional (3D) bioprinted matrices of varied stiffness. Cell-laden hydrogels were 3D bioprinted from gelatin methacryloyl (GelMA) using a digital light processing (DLP)-based 3D bioprinter operated with visible light (405 nm). Mechanical characterization revealed that the hydrogels had pathophysiologically relevant stiffnesses for a lymph node-mimetic tissue construct. EL4, a mouse T cell lymphoma line, or primary mouse T cells were 3D bioprinted and activated using a combination of 10 ng/mL of phorbol myristate acetate (PMA) and 0.1 μM of ionomycin. Cellular responses revealed differences between 2D and 3D cultures and that the biomechanical properties of the 3D bioprinted hydrogel influence T cell activation. Cellular responses of the 2D and 3D cultures in a soft matrix (19.83 ± 2.36 kPa) were comparable; however, they differed in a stiff matrix (52.95 ± 1.36 kPa). The fraction of viable EL4 cells was 1.3-fold higher in the soft matrix than in the stiff matrix. Furthermore, primary mouse T cells activated with PMA and ionomycin showed 1.35-fold higher viable cells in the soft matrix than in the stiff matrix. T cells bioprinted in a soft matrix and a stiff matrix released 7.4-fold and 5.9-fold higher amounts of interleukin-2 (IL-2) than 2D cultured cells, respectively. Overall, the study demonstrates the changes in the response of T cells in 3D bioprinted scaffolds toward engineering an ex vivo lymphoid tissue-mimetic system that can faithfully recapitulate T cell activation and unravel pathophysiological characteristics of T cells in infectious biology, autoimmunity, and cancers.
Collapse
Affiliation(s)
- Joel P Joseph
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru - 560012, India
| | | | - Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bengaluru - 560012, India
| | - Kaushik Chatterjee
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru - 560012, India
- Department of Materials Engineering, Indian Institute of Science, Bengaluru - 560012, India
| |
Collapse
|
49
|
Strating E, Verhagen MP, Wensink E, Dünnebach E, Wijler L, Aranguren I, De la Cruz AS, Peters NA, Hageman JH, van der Net MMC, van Schelven S, Laoukili J, Fodde R, Roodhart J, Nierkens S, Snippert H, Gloerich M, Rinkes IB, Elias SG, Kranenburg O. Co-cultures of colon cancer cells and cancer-associated fibroblasts recapitulate the aggressive features of mesenchymal-like colon cancer. Front Immunol 2023; 14:1053920. [PMID: 37261365 PMCID: PMC10228738 DOI: 10.3389/fimmu.2023.1053920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 05/03/2023] [Indexed: 06/02/2023] Open
Abstract
Background Poor prognosis in colon cancer is associated with a high content of cancer-associated fibroblasts (CAFs) and an immunosuppressive tumor microenvironment. The relationship between these two features is incompletely understood. Here, we aimed to generate a model system for studying the interaction between cancer cells and CAFs and their effect on immune-related cytokines and T cell proliferation. Methods CAFs were isolated from colon cancer liver metastases and were immortalized to prolong lifespan and improve robustness and reproducibility. Established medium and matrix compositions that support the growth of patient-derived organoids were adapted to also support CAF growth. Changes in growth pattern and cellular re-organization were assessed by confocal microscopy, live cell imaging, and immunofluorescence. Single cell RNA sequencing was used to study CAF/organoid co-culture-induced phenotypic changes in both cell types. Conditioned media were used to quantify the production of immunosuppressive factors and to assess their effect on T cell proliferation. Results We developed a co-culture system in which colon cancer organoids and CAFs spontaneously organize into superstructures with a high capacity to contract and stiffen the extracellular matrix (ECM). CAF-produced collagen IV provided a basement membrane supporting cancer cell organization into glandular structures, reminiscent of human cancer histology. Single cell RNA sequencing analysis showed that CAFs induced a partial epithelial-to-mesenchymal-transition in a subpopulation of cancer cells, similar to what is observed in the mesenchymal-like consensus molecular subtype 4 (CMS4) colon cancer. CAFs in co-culture were characterized by high expression of ECM components, ECM-remodeling enzymes, glycolysis, hypoxia, and genes involved in immunosuppression. An expression signature derived from CAFs in co-culture identified a subpopulation of glycolytic myofibroblasts specifically residing in CMS1 and CMS4 colon cancer. Medium conditioned by co-cultures contained high levels of the immunosuppressive factors TGFβ1, VEGFA and lactate, and potently inhibited T cell proliferation. Conclusion Co-cultures of organoids and immortalized CAFs recapitulate the histological, biophysical, and immunosuppressive features of aggressive mesenchymal-like human CRC. The model can be used to study the mechanisms of immunosuppression and to test therapeutic strategies targeting the cross-talk between CAFs and cancer cells. It can be further modified to represent distinct colon cancer subtypes and (organ-specific) microenvironments.
Collapse
Affiliation(s)
- Esther Strating
- Laboratory Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Emerens Wensink
- Department of Medical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ester Dünnebach
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Liza Wijler
- Laboratory Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Itziar Aranguren
- Laboratory Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Alberto Sanchez De la Cruz
- Laboratory Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Niek A. Peters
- Laboratory Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Joris H. Hageman
- Center for Molecular Medicine, Division LAB, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mirjam M. C. van der Net
- Center for Molecular Medicine, Division LAB, University Medical Center Utrecht, Utrecht, Netherlands
| | - Susanne van Schelven
- Laboratory Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jamila Laoukili
- Laboratory Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Riccardo Fodde
- Department of Pathology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Jeanine Roodhart
- Department of Medical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Stefan Nierkens
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Hugo Snippert
- Center for Molecular Medicine, Division LAB, University Medical Center Utrecht, Utrecht, Netherlands
| | - Martijn Gloerich
- Center for Molecular Medicine, Division LAB, University Medical Center Utrecht, Utrecht, Netherlands
| | - Inne Borel Rinkes
- Laboratory Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sjoerd G. Elias
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Onno Kranenburg
- Laboratory Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
- Utrecht Platform for Organoid Technology, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
50
|
Hyun J, Kim SJ, Cho SD, Kim HW. Mechano-modulation of T cells for cancer immunotherapy. Biomaterials 2023; 297:122101. [PMID: 37023528 DOI: 10.1016/j.biomaterials.2023.122101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 03/12/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023]
Abstract
Immunotherapy, despite its promise for future anti-cancer approach, faces significant challenges, such as off-tumor side effects, innate or acquired resistance, and limited infiltration of immune cells into stiffened extracellular matrix (ECM). Recent studies have highlighted the importance of mechano-modulation/-activation of immune cells (mainly T cells) for effective caner immunotherapy. Immune cells are highly sensitive to the applied physical forces and matrix mechanics, and reciprocally shape the tumor microenvironment. Engineering T cells with tuned properties of materials (e.g., chemistry, topography, and stiffness) can improve their expansion and activation ex vivo, and their ability to mechano-sensing the tumor specific ECM in vivo where they perform cytotoxic effects. T cells can also be exploited to secrete enzymes that soften ECM, thus increasing tumor infiltration and cellular therapies. Furthermore, T cells, such as chimeric antigen receptor (CAR)-T cells, genomic engineered to be spatiotemporally controllable by physical stimuli (e.g., ultrasound, heat, or light), can mitigate adverse off-tumor effects. In this review, we communicate these recent cutting-edge endeavors devoted to mechano-modulating/-activating T cells for effective cancer immunotherapy, and discuss future prospects and challenges in this field.
Collapse
|