1
|
Li Y, Yu Y, Xia B, Zhao S, Li X, Hu Q, Tian Y, Wang Y, Zhou Y, Yang C, Zhang D, Zhang Z, Kong L. Engineered vesicular cancer vaccines for immunosuppressive microenvironment reversion and in situ vaccine generation. J Control Release 2025; 382:113658. [PMID: 40122241 DOI: 10.1016/j.jconrel.2025.113658] [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: 12/10/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
It is crucial to reverse immunosuppressive tumor microenvironment (TME) and effectively activate both cellular and humoral immunity in cancer immunotherapy. We have found that decitabine, an epigenetic regulator, can increase antigen exposure and induce double-stranded RNA (dsRNA) accumulation in tumor cells. The corresponding cell-derived nanovesicles (NV) have the ability to stimulate both cellular and humoral immunity due to the internal dsRNA. However, the efficacy of dsRNA-containing NV (dsRNA@NV) remains constrained by the inadequate activation efficiency of immune cells in immunosuppressive TME. In this study, CD40L, an immune cell regulator, was incorporated on the surface of dsRNA@NV (dsRNA@NVCD40L) through lentiviral transfection to further reverse the immunosuppressive TME, by activating dendritic cells and regulating macrophages phenotypes via CD40-CD40L interaction. In addition, CD40L could induce immunogenic death of tumor cells, and the administration of dsRNA@NVCD40L effectively elicited an in situ cancer vaccine response in B16-OVA tumor. This proposed NV-based vaccine was expected to solve the problems of low immunogenicity, insufficient activation of immune responses and lack of effective regulation of immunosuppressive TME of existing tumor vesicular vaccines.
Collapse
Affiliation(s)
- Yang Li
- Department of Pharmaceutics, Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yulin Yu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bin Xia
- Graduate School of Human and Environmental Studies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Siyu Zhao
- Department of Pharmaceutics, Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaonan Li
- Department of Pharmaceutics, Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Hu
- Department of Pharmaceutics, Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yinmei Tian
- Department of Pharmaceutics, Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yi Wang
- Department of Pharmaceutics, Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yixuan Zhou
- Department of Pharmaceutics, Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Conglian Yang
- Department of Pharmaceutics, Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dan Zhang
- Department of Pharmacy, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China.
| | - Zhiping Zhang
- Department of Pharmaceutics, Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Engineering Research Centre for Novel Drug Delivery System, Wuhan 430030, China.
| | - Li Kong
- Department of Pharmaceutics, Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Engineering Research Centre for Novel Drug Delivery System, Wuhan 430030, China.
| |
Collapse
|
2
|
Zhu Y, Su Y, Guo Y, Wang X, Zhang Z, Lu Y, Yang H, Pang H. Current state of cancer immunity cycle: new strategies and challenges of using precision hydrogels to treat breast cancer. Front Immunol 2025; 16:1535464. [PMID: 40124373 PMCID: PMC11926806 DOI: 10.3389/fimmu.2025.1535464] [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: 11/27/2024] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
The cancer-immunity cycle provides a framework for a series of events in anti-cancer immune responses, initiated by T cell-mediated tumor cell killing, which leads to antigen presentation and T cell stimulation. Current immunomodulatory therapies for breast cancer are often associated with short duration, poor targeting to sites of action, and severe side effects. Hydrogels, with their extracellular matrix-mimicking properties, tunable characteristics, and diverse bioactivities, have garnered significant attention for their ability to locally deliver immunomodulators and cells, providing an immunomodulatory microenvironment to recruit, activate, and expand host immune cells. This review focuses on the design considerations of hydrogel platforms, including polymer backbone, crosslinking mechanisms, physicochemical properties, and immunomodulatory components. The immunomodulatory effects and therapeutic outcomes of various hydrogel systems in breast cancer treatment and tissue regeneration are highlighted, encompassing hydrogel depots for immunomodulator delivery, hydrogel scaffolds for cell delivery, and immunomodulatory hydrogels dependent on inherent material properties. Finally, the challenges that persist in current systems and future directions for immunomodulatory hydrogels are discussed.
Collapse
Affiliation(s)
- Yingze Zhu
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yanlin Su
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yaxin Guo
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xinyue Wang
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zhuoqi Zhang
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yige Lu
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hang Yang
- Department of Gastroenterology and Hepatology, Tianjin Second People’s Hospital, Tianjin, China
| | - Hui Pang
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
3
|
Pompano RR. Boosting B cells in blood-derived organoids. NATURE MATERIALS 2025; 24:173-175. [PMID: 39856416 DOI: 10.1038/s41563-024-02113-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
Affiliation(s)
- Rebecca R Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA.
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.
- Beirne B. Carter Immunology Center, University of Virginia, Charlottesville, VA, USA.
- Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
4
|
Britto LS, Balasubramani D, Desai S, Phillips P, Trehan N, Cesarman E, Koff JL, Singh A. T Cells Spatially Regulate B Cell Receptor Signaling in Lymphomas through H3K9me3 Modifications. Adv Healthc Mater 2025; 14:e2401192. [PMID: 38837879 PMCID: PMC11617604 DOI: 10.1002/adhm.202401192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/27/2024] [Indexed: 06/07/2024]
Abstract
Activated B cell-like diffuse large B-cell lymphoma (ABC-DLBCL) is a subtype associated with poor survival outcomes. Despite identifying therapeutic targets through molecular characterization, targeted therapies have limited success. New strategies using immune-competent tissue models are needed to understand how DLBCL cells evade treatment. Here, synthetic hydrogel-based lymphoma organoids are used to demonstrate how signals in the lymphoid tumor microenvironment (Ly-TME) can alter B cell receptor (BCR) signaling and specific histone modifications, tri-methylation of histone 3 at lysine 9 (H3K9me3), dampening the effects of BCR pathway inhibition. Using imaging modalities, T cells increase DNA methyltransferase 3A expression and cytoskeleton formation in proximal ABC-DLBCL cells, regulated by H3K9me3. Expansion microscopy on lymphoma organoids reveals T cells increase the size and quantity of segregated H3K9me3 clusters in ABC-DLBCL cells. Findings suggest the re-organization of higher-order chromatin structures that may contribute to evasion or resistance to therapy via the emergence of novel transcriptional states. Treating ABC-DLBCL cells with a G9α histone methyltransferase inhibitor reverses T cell-mediated modulation of H3K9me3 and overcomes T cell-mediated attenuation of treatment response to BCR pathway inhibition. This study emphasizes the Ly-TME's role in altering DLBCL fate and suggests targeting aberrant signaling and microenvironmental cross-talk that can benefit high-risk patients.
Collapse
Affiliation(s)
- Lucy S. Britto
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Deepali Balasubramani
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Sona Desai
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Phunterion Phillips
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Neev Trehan
- St Richards HospitalUniversity Hospitals Sussex NHS Foundation TrustChichesterWest SussexPO19 6SEUK
| | - Ethel Cesarman
- Department of Pathology and Laboratory MedicineWeill Cornell MedicineNew YorkNY10065USA
| | - Jean L. Koff
- Winship Cancer CenterEmory University School of MedicineAtlantaGA30307USA
| | - Ankur Singh
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
- Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30318USA
- Petit Institute for Bioengineering and BiosciencesGeorgia Institute of TechnologyAtlantaGA30332USA
| |
Collapse
|
5
|
Zhong Z, Quiñones-Pérez M, Dai Z, Juarez VM, Bhatia E, Carlson CR, Shah SB, Patel A, Fang Z, Hu T, Allam M, Hicks SL, Gupta M, Gupta SL, Weeks E, Vagelos SD, Molina A, Mulero-Russe A, Mora-Boza A, Joshi DJ, Sekaly RP, Sulchek T, Goudy SL, Wrammert J, Roy K, Boss JM, Coskun AF, Scharer CD, García AJ, Koff JL, Singh A. Human immune organoids to decode B cell response in healthy donors and patients with lymphoma. NATURE MATERIALS 2025; 24:297-311. [PMID: 39506098 PMCID: PMC11866935 DOI: 10.1038/s41563-024-02037-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/30/2024] [Indexed: 11/08/2024]
Abstract
Antibodies are produced when naive B cells differentiate into plasma cells within germinal centres (GCs) of lymphoid tissues. Patients with B cell lymphoma on effective immunotherapies exhibit diminished antibody production, leading to higher infection rates and reduced vaccine efficacy, even after B cell recovery. Current ex vivo models fail to sustain long-term GC reactions and effectively test B cell responses. Here we developed synthetic hydrogels mimicking the lymphoid tissue microenvironment, enabling human GCs from tonsils and peripheral blood mononuclear cell-derived B cells. Immune organoids derived from peripheral blood mononuclear cells maintain GC B cells and plasma cells longer than tonsil-derived ones and exhibit unique B cell programming, including GC compartments, somatic hypermutation, immunoglobulin class switching and B cell clones. Chemical inhibition of transcriptional and epigenetic processes enhances plasma cell formation. While integrating polarized CXCL12 protein in a lymphoid organ-on-chip modulates GC responses in healthy donor B cells, it fails with B cells derived from patients with lymphoma. Our system allows rapid, controlled modelling of immune responses and B cell disorders.
Collapse
Affiliation(s)
- Zhe Zhong
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Manuel Quiñones-Pérez
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhonghao Dai
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Valeria M Juarez
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Eshant Bhatia
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Christopher R Carlson
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shivem B Shah
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Anjali Patel
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhou Fang
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Thomas Hu
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Mayar Allam
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sakeenah L Hicks
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Mansi Gupta
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Sneh Lata Gupta
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Ethan Weeks
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Stephanie D Vagelos
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Alejandro Molina
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Adriana Mulero-Russe
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ana Mora-Boza
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Devyani J Joshi
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Rafick P Sekaly
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Todd Sulchek
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Steven L Goudy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Otolaryngology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Jens Wrammert
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Krishnendu Roy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jean L Koff
- Winship Cancer Center, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Ankur Singh
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
6
|
Cook SR, Ball AG, Mohammad A, Pompano RR. A 3D-printed multi-compartment organ-on-chip platform with a tubing-free pump models communication with the lymph node. LAB ON A CHIP 2025; 25:155-174. [PMID: 39661075 PMCID: PMC11633827 DOI: 10.1039/d4lc00489b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
Multi-organ-on-chip systems (MOOCs) have the potential to mimic communication between organ systems and reveal mechanisms of health and disease. However, many existing MOOCs are challenging for non-experts to implement due to complex tubing, electronics, or pump mechanisms. In addition, few MOOCs have incorporated immune organs such as the lymph node (LN), limiting their applicability to model critical events such as vaccination. Here we developed a 3D-printed, user-friendly device and companion tubing-free impeller pump with the capacity to co-culture two or more tissue samples, including a LN, under a recirculating common media. Native tissue structure and immune function were incorporated by maintaining slices of murine LN tissue ex vivo in 3D-printed mesh supports for at least 24 h. In a two-compartment model of a LN and an upstream injection site in mock tissue, vaccination of the multi-compartment chip was similar to in vivo vaccination in terms of locations of antigen accumulation and acute changes in activation markers and gene expression in the LN. We anticipate that in the future, this flexible platform will enable models of multi-organ immune responses throughout the body.
Collapse
Affiliation(s)
- Sophie R Cook
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA.
| | - Alexander G Ball
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Carter Immunology Center and UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, USA
| | | | - Rebecca R Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA.
- Carter Immunology Center and UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
7
|
Yu Z, Dong C, Yang Y, Zheng Z, Ge X. USP21 stabilizes immune checkpoint of CD276 and serves as an immunological and tumor prognostic biomarker. Biochem Biophys Res Commun 2024; 745:151221. [PMID: 39736236 DOI: 10.1016/j.bbrc.2024.151221] [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: 10/18/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/01/2025]
Abstract
Ubiquitin-specific protease 21 (USP21) belongs to the ubiquitin-specific protease family and is a member of the deubiquitinating enzyme (DUB) family. Previous research has shown that USP21 promotes cancer initiation and progression. However, there have been few pan-cancer analysis on USP21. We analyzed the expression levels of USP21 mRNA and protein in various human tumor tissues using several public databases such as The Cancer Genome Atlas (TCGA), Genotype Tissue Expression (GTEx), and Human Protein Atlas (HPA). Kaplan-Meier survival analyses were utilized to test the effect of USP21 on overall survival (OS) and progression-free interval (PFS) of these tumor patients. Our study demonstrated that USP21 was differentially expressed between normal and malignant tissues, conferring a notable value in evaluation of prognosis and diagnosis. In addition, enrichment and correlation analyses linking USP21 with immune features such as immune-cell-infiltration rate and immune-checkpoint-gene expression indicated that USP21 is an applicable immunotherapeutic marker for liver cancer. To further elucidate the role of USP21, we downregulated its expression in hepatocellular carcinoma cells and identified a remarkable decrease in expression of the immune checkpoint CD276, which contributes to the immune escape of tumor cells by suppressing the immune system. Together, our results indicated a promising potential of USP21 for future tumor prevention.
Collapse
Affiliation(s)
- Zhu Yu
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Chengyuan Dong
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Yanrong Yang
- Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China
| | - Zening Zheng
- Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China
| | - Xin Ge
- Department of Clinical Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
8
|
Santamaria-Martínez A, Epiney J, Srivastava D, Tavernari D, Varrone M, Milowich D, Letovanec I, Krueger T, Duran R, Ciriello G, Cairoli A, Oricchio E. Development of patient-derived lymphomoids with preserved tumor architecture for lymphoma therapy screening. Nat Commun 2024; 15:10650. [PMID: 39653701 PMCID: PMC11628617 DOI: 10.1038/s41467-024-55098-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024] Open
Abstract
The efficacy of anti-cancer therapies depends on the genomic composition of the tumor, its microenvironment, spatial organization, and intra-tumor heterogeneity. B-cell lymphomas are a heterogeneous group of tumors emerging from B-cells at different stages of differentiation and exhibiting tumor-specific interactions with the tumor microenvironment. Thus, the effect of drug treatments can be influenced by the tumor composition and functional interactions among immune cells. Here, we develop a platform to maintain small fragments of human lymphoma tissue in culture for several days, and use them to test response to small molecules. We collect 27 patient samples representative of different lymphoma subtypes, and establish ex vivo tissue fragments that retain histological, cellular, and molecular characteristics of the original tissue, here referred to as lymphomoids. Using lymphomoids, we test sensitivity to several clinically approved drugs in parallel and examine tissue remodeling upon treatment. Moreover, when this information is available, we show that the effect of the inhibitors observed in lymphomoids is consistent with the patients' response in the clinic. Thus, lymphomoids represent an innovative ex vivo model to assess the effect of anti-cancer therapies while preserving the tissue structure and its components.
Collapse
Affiliation(s)
- Albert Santamaria-Martínez
- Swiss Institute for Experimental Cancer Research, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Swiss Cancer Center Léman, Lausanne, Switzerland.
| | - Justine Epiney
- Swiss Institute for Experimental Cancer Research, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Swiss Cancer Center Léman, Lausanne, Switzerland
- Division of Hematology and Central Hematology Laboratory, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Divyanshu Srivastava
- Swiss Institute for Experimental Cancer Research, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Swiss Cancer Center Léman, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Daniele Tavernari
- Swiss Institute for Experimental Cancer Research, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Swiss Cancer Center Léman, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Marco Varrone
- Swiss Cancer Center Léman, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Dina Milowich
- Institut Central des Hôpitaux (ICH), Hôpital du Valais, Sion, Switzerland
| | - Igor Letovanec
- Institut Central des Hôpitaux (ICH), Hôpital du Valais, Sion, Switzerland
| | | | - Rafael Duran
- Department of Diagnostic and Interventional Radiology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Giovanni Ciriello
- Swiss Cancer Center Léman, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Anne Cairoli
- Division of Hematology and Central Hematology Laboratory, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Elisa Oricchio
- Swiss Institute for Experimental Cancer Research, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Swiss Cancer Center Léman, Lausanne, Switzerland.
| |
Collapse
|
9
|
Polak R, Zhang ET, Kuo CJ. Cancer organoids 2.0: modelling the complexity of the tumour immune microenvironment. Nat Rev Cancer 2024; 24:523-539. [PMID: 38977835 DOI: 10.1038/s41568-024-00706-6] [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: 05/09/2024] [Indexed: 07/10/2024]
Abstract
The development of neoplasia involves a complex and continuous interplay between malignantly transformed cells and the tumour microenvironment (TME). Cancer immunotherapies targeting the immune TME have been increasingly validated in clinical trials but response rates vary substantially between tumour histologies and are often transient, idiosyncratic and confounded by resistance. Faithful experimental models of the patient-specific tumour immune microenvironment, capable of recapitulating tumour biology and immunotherapy effects, would greatly improve patient selection, target identification and definition of resistance mechanisms for immuno-oncology therapeutics. In this Review, we discuss currently available and rapidly evolving 3D tumour organoid models that capture important immune features of the TME. We highlight diverse opportunities for organoid-based investigations of tumour immunity, drug development and precision medicine.
Collapse
Affiliation(s)
- Roel Polak
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Elisa T Zhang
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
10
|
Yee Mon KJ, Kim S, Dai Z, West JD, Zhu H, Jain R, Grimson A, Rudd BD, Singh A. Functionalized nanowires for miRNA-mediated therapeutic programming of naïve T cells. NATURE NANOTECHNOLOGY 2024; 19:1190-1202. [PMID: 38684809 PMCID: PMC11330359 DOI: 10.1038/s41565-024-01649-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/13/2024] [Indexed: 05/02/2024]
Abstract
Cellular programming of naïve T cells can improve the efficacy of adoptive T-cell therapy. However, the current ex vivo engineering of T cells requires the pre-activation of T cells, which causes them to lose their naïve state. In this study, cationic-polymer-functionalized nanowires were used to pre-program the fate of primary naïve CD8+ T cells to achieve a therapeutic response in vivo. This was done by delivering single or multiple microRNAs to primary naïve mouse and human CD8+ T cells without pre-activation. The use of nanowires further allowed for the delivery of large, whole lentiviral particles with potential for long-term integration. The combination of deletion and overexpression of miR-29 and miR-130 impacted the ex vivo T-cell differentiation fate from the naïve state. The programming of CD8+ T cells using nanowire-delivered co-delivery of microRNAs resulted in the modulation of T-cell fitness by altering the T-cell proliferation, phenotypic and transcriptional regulation, and secretion of effector molecules. Moreover, the in vivo adoptive transfer of murine CD8+ T cells programmed through the nanowire-mediated dual delivery of microRNAs provided enhanced immune protection against different types of intracellular pathogen (influenza and Listeria monocytogenes). In vivo analyses demonstrated that the simultaneous alteration of miR-29 and miR-130 levels in naïve CD8+ T cells reduces the persistence of canonical memory T cells whereas increases the population of short-lived effector T cells. Nanowires could potentially be used to modulate CD8+ T-cell differentiation and achieve a therapeutic response in vivo without the need for pre-activation.
Collapse
Affiliation(s)
- Kristel J Yee Mon
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Sungwoong Kim
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA
| | - Zhonghao Dai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Jessica D West
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY, USA
| | - Hongya Zhu
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY, USA
| | - Ritika Jain
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Andrew Grimson
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY, USA
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA.
| | - Ankur Singh
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
11
|
Zhang Y, Chu J, Hou Q, Qian S, Wang Z, Yang Q, Song W, Dong L, Shi Z, Gao Y, Meng M, Zhang M, Zhang X, Chen Q. Ageing microenvironment mediates lymphocyte carcinogenesis and lymphoma drug resistance: From mechanisms to clinical therapy (Review). Int J Oncol 2024; 64:65. [PMID: 38757347 PMCID: PMC11095602 DOI: 10.3892/ijo.2024.5653] [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: 12/13/2023] [Accepted: 04/08/2024] [Indexed: 05/18/2024] Open
Abstract
Cellular senescence has a complex role in lymphocyte carcinogenesis and drug resistance of lymphomas. Senescent lymphoma cells combine with immunocytes to create an ageing environment that can be reprogrammed with a senescence‑associated secretory phenotype, which gradually promotes therapeutic resistance. Certain signalling pathways, such as the NF‑κB, Wnt and PI3K/AKT/mTOR pathways, regulate the tumour ageing microenvironment and induce the proliferation and progression of lymphoma cells. Therefore, targeting senescence‑related enzymes or their signal transduction pathways may overcome radiotherapy or chemotherapy resistance and enhance the efficacy of relapsed/refractory lymphoma treatments. Mechanisms underlying drug resistance in lymphomas are complex. The ageing microenvironment is a novel factor that contributes to drug resistance in lymphomas. In terms of clinical translation, some senolytics have been used in clinical trials on patients with relapsed or refractory lymphoma. Combining immunotherapy with epigenetic drugs may achieve better therapeutic effects; however, senescent cells exhibit considerable heterogeneity and lymphoma has several subtypes. Extensive research is necessary to achieve the practical application of senolytics in relapsed or refractory lymphomas. This review summarises the mechanisms of senescence‑associated drug resistance in lymphoma, as well as emerging strategies using senolytics, to overcome therapeutic resistance in lymphoma.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jingwen Chu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Qi Hou
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Siyu Qian
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zeyuan Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Qing Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Wenting Song
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ling Dong
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhuangzhuang Shi
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yuyang Gao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Miaomiao Meng
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Qingjiang Chen
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
12
|
Huayamares SG, Loughrey D, Kim H, Dahlman JE, Sorscher EJ. Nucleic acid-based drugs for patients with solid tumours. Nat Rev Clin Oncol 2024; 21:407-427. [PMID: 38589512 DOI: 10.1038/s41571-024-00883-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/10/2024]
Abstract
The treatment of patients with advanced-stage solid tumours typically involves a multimodality approach (including surgery, chemotherapy, radiotherapy, targeted therapy and/or immunotherapy), which is often ultimately ineffective. Nucleic acid-based drugs, either as monotherapies or in combination with standard-of-care therapies, are rapidly emerging as novel treatments capable of generating responses in otherwise refractory tumours. These therapies include those using viral vectors (also referred to as gene therapies), several of which have now been approved by regulatory agencies, and nanoparticles containing mRNAs and a range of other nucleotides. In this Review, we describe the development and clinical activity of viral and non-viral nucleic acid-based treatments, including their mechanisms of action, tolerability and available efficacy data from patients with solid tumours. We also describe the effects of the tumour microenvironment on drug delivery for both systemically administered and locally administered agents. Finally, we discuss important trends resulting from ongoing clinical trials and preclinical testing, and manufacturing and/or stability considerations that are expected to underpin the next generation of nucleic acid agents for patients with solid tumours.
Collapse
Affiliation(s)
- Sebastian G Huayamares
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| | - David Loughrey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| | - Hyejin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Emory University School of Medicine, Atlanta, GA, USA.
| | - Eric J Sorscher
- Emory University School of Medicine, Atlanta, GA, USA.
- Department of Pediatrics, Emory University, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
13
|
Gewurz B, Guo R, Lim M, Shah H, Paulo J, Zhang Y, Yang H, Wang LW, Strebinger D, Smith N, Li M, Leong M, Lutchenkov M, Liang JH, Li Z, Wang Y, Puri R, Melnick A, Green M, Asara J, Papathanassiu A, Gygi S, Mootha V. Multi-omic Analysis of Human B-cell Activation Reveals a Key Lysosomal BCAT1 Role in mTOR Hyperactivation by B-cell receptor and TLR9. RESEARCH SQUARE 2024:rs.3.rs-4413958. [PMID: 38854072 PMCID: PMC11160916 DOI: 10.21203/rs.3.rs-4413958/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
B-lymphocytes play major adaptive immune roles, producing antibody and driving T-cell responses. However, how immunometabolism networks support B-cell activation and differentiation in response to distinct receptor stimuli remains incompletely understood. To gain insights, we systematically investigated acute primary human B-cell transcriptional, translational and metabolomic responses to B-cell receptor (BCR), Toll-like receptor 9 (TLR9), CD40-ligand (CD40L), interleukin-4 (IL4) or combinations thereof. T-independent BCR/TLR9 co-stimulation, which drives malignant and autoimmune B-cell states, jointly induced PD-L1 plasma membrane expression, supported by NAD metabolism and oxidative phosphorylation. BCR/TLR9 also highly induced the transaminase BCAT1, which localized to lysosomal membranes to support branched chain amino acid synthesis and mTORC1 hyperactivation. BCAT1 inhibition blunted BCR/TLR9, but not CD40L/IL4-triggered B-cell proliferation, IL10 expression and BCR/TLR pathway-driven lymphoma xenograft outgrowth. These results provide a valuable resource, reveal receptor-mediated immunometabolism remodeling to support key B-cell phenotypes including PD-L1 checkpoint signaling, and identify BCAT1 as a novel B-cell therapeutic target.
Collapse
Affiliation(s)
| | | | - Matthew Lim
- Department of Cell Biology, Harvard Medical School
| | | | | | | | - Haopeng Yang
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center
| | | | | | | | - Meng Li
- Department of Medicine, Division of Hematology & Medical Oncology, Weill Cornell Medicine
| | | | | | | | | | | | - Rishi Puri
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University
| | | | - Michael Green
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center
| | | | | | | | | |
Collapse
|
14
|
Zareein A, Mahmoudi M, Jadhav SS, Wilmore J, Wu Y. Biomaterial engineering strategies for B cell immunity modulations. Biomater Sci 2024; 12:1981-2006. [PMID: 38456305 PMCID: PMC11019864 DOI: 10.1039/d3bm01841e] [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: 11/11/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
B cell immunity has a penetrating effect on human health and diseases. Therapeutics aiming to modulate B cell immunity have achieved remarkable success in combating infections, autoimmunity, and malignancies. However, current treatments still face significant limitations in generating effective long-lasting therapeutic B cell responses for many conditions. As the understanding of B cell biology has deepened in recent years, clearer regulation networks for B cell differentiation and antibody production have emerged, presenting opportunities to overcome current difficulties and realize the full therapeutic potential of B cell immunity. Biomaterial platforms have been developed to leverage these emerging concepts to augment therapeutic humoral immunity by facilitating immunogenic reagent trafficking, regulating T cell responses, and modulating the immune microenvironment. Moreover, biomaterial engineering tools have also advanced our understanding of B cell biology, further expediting the development of novel therapeutics. In this review, we will introduce the general concept of B cell immunobiology and highlight key biomaterial engineering strategies in the areas including B cell targeted antigen delivery, sustained B cell antigen delivery, antigen engineering, T cell help optimization, and B cell suppression. We will also discuss our perspective on future biomaterial engineering opportunities to leverage humoral immunity for therapeutics.
Collapse
Affiliation(s)
- Ali Zareein
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Mina Mahmoudi
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Shruti Sunil Jadhav
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
| | - Joel Wilmore
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
15
|
Kastenschmidt JM, Schroers-Martin JG, Sworder BJ, Sureshchandra S, Khodadoust MS, Liu CL, Olsen M, Kurtz DM, Diehn M, Wagar LE, Alizadeh AA. A human lymphoma organoid model for evaluating and targeting the follicular lymphoma tumor immune microenvironment. Cell Stem Cell 2024; 31:410-420.e4. [PMID: 38402619 PMCID: PMC10960522 DOI: 10.1016/j.stem.2024.01.012] [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: 09/01/2023] [Revised: 12/11/2023] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
Heterogeneity in the tumor microenvironment (TME) of follicular lymphomas (FLs) can affect clinical outcomes. Current immunotherapeutic strategies, including antibody- and cell-based therapies, variably overcome pro-tumorigenic mechanisms for sustained disease control. Modeling the intact FL TME, with its native, syngeneic tumor-infiltrating leukocytes, is a major challenge. Here, we describe an organoid culture method for cultivating patient-derived lymphoma organoids (PDLOs), which include cells from the native FL TME. We define the robustness of this method by successfully culturing cryopreserved FL specimens from diverse patients and demonstrate the stability of TME cellular composition, tumor somatic mutations, gene expression profiles, and B/T cell receptor dynamics over 3 weeks. PDLOs treated with CD3:CD19 and CD3:CD20 therapeutic bispecific antibodies showed B cell killing and T cell activation. This stable system offers a robust platform for advancing precision medicine efforts in FL through patient-specific modeling, high-throughput screening, TME signature identification, and treatment response evaluation.
Collapse
Affiliation(s)
- Jenna M Kastenschmidt
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92617, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92617, USA; Cancer Research Institute, University of California, Irvine, Irvine, CA 92617, USA
| | | | - Brian J Sworder
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Suhas Sureshchandra
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92617, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92617, USA; Cancer Research Institute, University of California, Irvine, Irvine, CA 92617, USA
| | - Michael S Khodadoust
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA
| | - Chih Long Liu
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mari Olsen
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - David M Kurtz
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Lisa E Wagar
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92617, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92617, USA; Cancer Research Institute, University of California, Irvine, Irvine, CA 92617, USA.
| | - Ash A Alizadeh
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Division of Hematology, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
16
|
Wheeler TA, Antoinette AY, Bhatia E, Kim MJ, Ijomanta CN, Zhao A, van der Meulen MCH, Singh A. Mechanical loading of joint modulates T cells in lymph nodes to regulate osteoarthritis. Osteoarthritis Cartilage 2024; 32:287-298. [PMID: 38072172 PMCID: PMC10955501 DOI: 10.1016/j.joca.2023.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023]
Abstract
OBJECTIVE The crosstalk of joint pathology with local lymph nodes in osteoarthritis (OA) is poorly understood. We characterized the change in T cells in lymph nodes following load-induced OA and established the association of the presence and migration of T cells to the onset and progression of OA. METHODS We used an in vivo model of OA to induce mechanical load-induced joint damage. After cyclic tibial compression of mice, we analyzed lymph nodes for T cells using flow cytometry and joint pathology using histology and microcomputed tomography. The role of T-cell migration and the presence of T-cell type was examined using T-cell receptor (TCR)α-/- mice and an immunomodulatory drug, Sphingosine-1-phosphate (S1P) receptor inhibitor-treated mice, respectively. RESULTS We demonstrated a significant increase in T-cell populations in local lymph nodes in response to joint injury in 10, 16, and 26-week-old mice, and as a function of load duration, 1, 2, and 6 weeks. T-cell expression of inflammatory cytokine markers increased in the local lymph nodes and was associated with load-induced OA progression in the mouse knee. Joint loading in TCRα-/- mice reduced both cartilage degeneration (Osteoarthritis Research Society International (OARSI) scores: TCRα 0.568, 0.981-0.329 confidence interval (CI); wild type (WT) 1.328, 2.353-0.749 CI) and osteophyte formation. Inhibition of T-cell egress from lymph nodes attenuated load-induced cartilage degradation (OARSI scores: Fingolimod: 0.509, 1.821-0.142 CI; Saline 1.210, 1.932-0.758 CI) and decreased localization of T cells in the synovium. CONCLUSIONS These results establish the association of lymph node-resident T cells in joint damage and suggest that the S1P receptor modulators and T-cell immunotherapies could be used to treat OA.
Collapse
Affiliation(s)
- Tibra A Wheeler
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Adrien Y Antoinette
- Sibley School of Mechanical & Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Eshant Bhatia
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Matthew J Kim
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Ann Zhao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Marjolein C H van der Meulen
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA; Sibley School of Mechanical & Aerospace Engineering, Cornell University, Ithaca, NY, USA; Research Division, Hospital for Special Surgery, New York, NY, USA.
| | - Ankur Singh
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA; Sibley School of Mechanical & Aerospace Engineering, Cornell University, Ithaca, NY, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA; Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
17
|
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
|
18
|
Zhou R, Guo J, Feng X, Zhou W. Mechanisms of the role of proto-oncogene activation in promoting malignant transformation of mature B cells. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:113-121. [PMID: 38615172 PMCID: PMC11017026 DOI: 10.11817/j.issn.1672-7347.2024.230304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Indexed: 04/15/2024]
Abstract
Malignant tumors continue to pose a significant threat to human life and safety and their development is primarily due to the activation of proto-oncogenes and the inactivation of suppressor genes. Among these, the activation of proto-oncogenes possesses greater potential to drive the malignant transformation of cells. Targeting oncogenes involved in the malignant transformation of tumor cells has provided a novel approach for the development of current antitumor drugs. Several preclinical and clinical studies have revealed that the development pathway of B cells, and the malignant transformation of mature B cells into tumors have been regulated by oncogenes and their metabolites. Therefore, summarizing the key oncogenes involved in the process of malignant transformation of mature B cells and elucidating the mechanisms of action in tumor development hold significant importance for the clinical treatment of malignant tumors.
Collapse
Affiliation(s)
- Ruiqi Zhou
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha 410078.
| | - Jiaojiao Guo
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008
| | - Xiangling Feng
- Xiangya School of Public Health, Central South University, Changsha 410006, China
| | - Wen Zhou
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha 410078.
| |
Collapse
|
19
|
Sullivan MR, White RP, Dashnamoorthy Ravi, Kanetkar N, Fridman IB, Ekenseair A, Evens AM, Konry T. Characterizing influence of rCHOP treatment on diffuse large B-cell lymphoma microenvironment through in vitro microfluidic spheroid model. Cell Death Dis 2024; 15:18. [PMID: 38195589 PMCID: PMC10776622 DOI: 10.1038/s41419-023-06299-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/11/2023] [Accepted: 11/13/2023] [Indexed: 01/11/2024]
Abstract
For over two decades, Rituximab and CHOP combination treatment (rCHOP) has remained the standard treatment approach for diffuse large B-cell lymphoma (DLBCL). Despite numerous clinical trials exploring treatment alternatives, few options have shown any promise at further improving patient survival and recovery rates. A wave of new therapeutic approaches have recently been in development with the rise of immunotherapy for cancer, however, the cost of clinical trials is prohibitive of testing all promising approaches. Improved methods of early drug screening are essential for expediting the development of the therapeutic approaches most likely to help patients. Microfluidic devices provide a powerful tool for drug testing with enhanced biological relevance, along with multi-parameter data outputs. Here, we describe a hydrogel spheroid-based microfluidic model for screening lymphoma treatments. We utilized primary patient DLBCL cells in combination with NK cells and rCHOP treatment to determine the biological relevance of this approach. We observed cellular viability in response to treatment, rheological properties, and cell surface marker expression levels correlated well with expected in vivo characteristics. In addition, we explored secretory and transcriptomic changes in response to treatment. Our results showed complex changes in phenotype and transcriptomic response to treatment stimuli, including numerous metabolic and immunogenic changes. These findings support this model as an optimal platform for the comparative screening of novel treatments.
Collapse
Affiliation(s)
- Matthew R Sullivan
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Rachel P White
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | | | - Ninad Kanetkar
- Chemical Engineering Department, Northeastern University, Boston, MA, USA
| | - Ilana Berger Fridman
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
- Avram and Stella Goldstein-Goren Department of Biotechnology and Regenerative Medicine and Stem Cell Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Adam Ekenseair
- Chemical Engineering Department, Northeastern University, Boston, MA, USA
| | | | - Tania Konry
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA.
| |
Collapse
|
20
|
Wang M, Phan S, Hayes BH, Discher DE. Genetic heterogeneity in p53-null leukemia increases transiently with spindle assembly checkpoint inhibition and is not rescued by p53. Chromosoma 2024; 133:77-92. [PMID: 37256347 PMCID: PMC10828900 DOI: 10.1007/s00412-023-00800-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/08/2023] [Accepted: 05/16/2023] [Indexed: 06/01/2023]
Abstract
Chromosome gains or losses often lead to copy number variations (CNV) and loss of heterozygosity (LOH). Both quantities are low in hematologic "liquid" cancers versus solid tumors in data of The Cancer Genome Atlas (TCGA) that also shows the fraction of a genome affected by LOH is ~ one-half of that with CNV. Suspension cultures of p53-null THP-1 leukemia-derived cells conform to these trends, despite novel evidence here of genetic heterogeneity and transiently elevated CNV after perturbation. Single-cell DNAseq indeed reveals at least 8 distinct THP-1 aneuploid clones with further intra-clonal variation, suggesting ongoing genetic evolution. Importantly, acute inhibition of the mitotic spindle assembly checkpoint (SAC) produces CNV levels that are typical of high-CNV solid tumors, with subsequent cell death and down-selection to novel CNV. Pan-cancer analyses show p53 inactivation associates with aneuploidy, but leukemias exhibit a weaker trend even though p53 inactivation correlates with poor survival. Overexpression of p53 in THP-1 does not rescue established aneuploidy or LOH but slightly increases cell death under oxidative or confinement stress, and triggers p21, a key p53 target, but without affecting net growth. Our results suggest that factors other than p53 exert stronger pressures against aneuploidy in liquid cancers, and identifying such CNV suppressors could be useful across liquid and solid tumor types.
Collapse
Affiliation(s)
- Mai Wang
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Steven Phan
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Brandon H Hayes
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dennis E Discher
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
21
|
Brauge B, Dessauge E, Creusat F, Tarte K. Modeling the crosstalk between malignant B cells and their microenvironment in B-cell lymphomas: challenges and opportunities. Front Immunol 2023; 14:1288110. [PMID: 38022603 PMCID: PMC10652758 DOI: 10.3389/fimmu.2023.1288110] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
B-cell lymphomas are a group of heterogeneous neoplasms resulting from the clonal expansion of mature B cells arrested at various stages of differentiation. Specifically, two lymphoma subtypes arise from germinal centers (GCs), namely follicular lymphoma (FL) and GC B-cell diffuse large B-cell lymphoma (GCB-DLBCL). In addition to recent advances in describing the genetic landscape of FL and GCB-DLBCL, tumor microenvironment (TME) has progressively emerged as a central determinant of early lymphomagenesis, subclonal evolution, and late progression/transformation. The lymphoma-supportive niche integrates a dynamic and coordinated network of immune and stromal cells defining microarchitecture and mechanical constraints and regulating tumor cell migration, survival, proliferation, and immune escape. Several questions are still unsolved regarding the interplay between lymphoma B cells and their TME, including the mechanisms supporting these bidirectional interactions, the impact of the kinetic and spatial heterogeneity of the tumor niche on B-cell heterogeneity, and how individual genetic alterations can trigger both B-cell intrinsic and B-cell extrinsic signals driving the reprogramming of non-malignant cells. Finally, it is not clear whether these interactions might promote resistance to treatment or, conversely, offer valuable therapeutic opportunities. A major challenge in addressing these questions is the lack of relevant models integrating tumor cells with specific genetic hits, non-malignant cells with adequate functional properties and organization, extracellular matrix, and biomechanical forces. We propose here an overview of the 3D in vitro models, xenograft approaches, and genetically-engineered mouse models recently developed to study GC B-cell lymphomas with a specific focus on the pros and cons of each strategy in understanding B-cell lymphomagenesis and evaluating new therapeutic strategies.
Collapse
Affiliation(s)
- Baptiste Brauge
- UMR 1236, Univ Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue, Rennes, France
| | - Elise Dessauge
- UMR 1236, Univ Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue, Rennes, France
| | - Florent Creusat
- UMR 1236, Univ Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue, Rennes, France
| | - Karin Tarte
- UMR 1236, Univ Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue, Rennes, France
- SITI Laboratory, Centre Hospitalier Universitaire (CHU) Rennes, Etablissement Français du sang, Univ Rennes, Rennes, France
| |
Collapse
|
22
|
Decombis S, Bellanger C, Le Bris Y, Madiot C, Jardine J, Santos JC, Boulet D, Dousset C, Menard A, Kervoelen C, Douillard E, Moreau P, Minvielle S, Moreau-Aubry A, Tessoulin B, Roue G, Bidère N, Le Gouill S, Pellat-Deceunynck C, Chiron D. CARD11 gain of function upregulates BCL2A1 expression and promotes resistance to targeted therapies combination in B-cell lymphoma. Blood 2023; 142:1543-1555. [PMID: 37562004 DOI: 10.1182/blood.2023020211] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/07/2023] [Accepted: 07/07/2023] [Indexed: 08/12/2023] Open
Abstract
A strategy combining targeted therapies is effective in B-cell lymphomas (BCL), such as mantle cell lymphoma (MCL), but acquired resistances remain a recurrent issue. In this study, we performed integrative longitudinal genomic and single-cell RNA-sequencing analyses of patients with MCL who were treated with targeted therapies against CD20, BCL2, and Bruton tyrosine kinase (OAsIs trial). We revealed the emergence of subclones with a selective advantage against OAsIs combination in vivo and showed that resistant cells were characterized by B-cell receptor (BCR)-independent overexpression of NF-κB1 target genes, especially owing to CARD11 mutations. Functional studies demonstrated that CARD11 gain of function not only resulted in BCR independence but also directly increased the transcription of the antiapoptotic BCL2A1, leading to resistance against venetoclax and OAsIs combination. Based on the transcriptional profile of OAsIs-resistant subclones, we designed a 16-gene resistance signature that was also predictive for patients with MCL who were treated with conventional chemotherapy, underlying a common escape mechanism. Among druggable strategies to inhibit CARD11-dependent NF-κB1 transduction, we evaluated the selective inhibition of its essential partner MALT1. We demonstrated that MALT1 protease inhibition led to a reduction in the expression of genes involved in OAsIs resistance, including BCL2A1. Consequently, MALT1 inhibition induced synergistic cell death in combination with BCL2 inhibition, irrespective of CARD11 mutational status, both in vitro and in vivo. Taken together, our study identified mechanisms of resistance to targeted therapies and provided a novel strategy to overcome resistance in aggressive BCL. The OAsIs trial was registered at www.clinicaltrials.gov #NCT02558816.
Collapse
Affiliation(s)
- Salomé Decombis
- Hematology Department, Nantes Université, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Celine Bellanger
- Hematology Department, Nantes Université, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Yannick Le Bris
- Hematology Department, Nantes Université, Centre Hospitalier Universitaire de Nantes, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Candice Madiot
- Hematology Department, Nantes Université, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Jane Jardine
- Hematology Department, Nantes Université, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | | | - Delphine Boulet
- Hematology Department, Nantes Université, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Christelle Dousset
- Hematology Department, Nantes Université, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Audrey Menard
- Hematology Department, Nantes Université, Centre Hospitalier Universitaire de Nantes, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Charlotte Kervoelen
- Therassay (Onco-Hemato) Core Facility, Nantes Université, Capacités, Nantes, France
| | - Elise Douillard
- Hematology Department, Nantes Université, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Philippe Moreau
- Hematology Department, Nantes Université, Centre Hospitalier Universitaire de Nantes, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Stephane Minvielle
- Hematology Department, Nantes Université, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Agnes Moreau-Aubry
- Hematology Department, Nantes Université, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Benoit Tessoulin
- Hematology Department, Nantes Université, Centre Hospitalier Universitaire de Nantes, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Gael Roue
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | - Nicolas Bidère
- Hematology Department, Nantes Université, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | | | | | - David Chiron
- Hematology Department, Nantes Université, INSERM, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| |
Collapse
|
23
|
Martin KE, Hunckler MD, Chee E, Caplin JD, Barber GF, Kalelkar PP, Schneider RS, García AJ. Hydrolytic hydrogels tune mesenchymal stem cell persistence and immunomodulation for enhanced diabetic cutaneous wound healing. Biomaterials 2023; 301:122256. [PMID: 37517209 PMCID: PMC10529272 DOI: 10.1016/j.biomaterials.2023.122256] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/20/2023] [Accepted: 07/23/2023] [Indexed: 08/01/2023]
Abstract
Diabetes is associated with an altered global inflammatory state with impaired wound healing. Mesenchymal stem/stromal cells (MSC) are being explored for treatment of diabetic cutaneous wounds due to their regenerative properties. These cells are commonly delivered by injection, but the need to prolong the retention of MSC at sites of injury has spurred the development of biomaterial-based MSC delivery vehicles. However, controlling biomaterial degradation rates in vivo remains a therapeutic-limiting challenge. Here, we utilize hydrolytically degradable ester linkages to engineer synthetic hydrogels with tunable in vivo degradation kinetics for temporally controlled delivery of MSC. In vivo hydrogel degradation rate can be controlled by altering the ratio of ester to amide linkages in the hydrogel macromers. These hydrolytic hydrogels degrade at rates that enable unencumbered cutaneous wound healing, while enhancing the local persistence MSC compared to widely used protease-degradable hydrogels. Furthermore, hydrogel-based delivery of MSC modulates local immune responses and enhances cutaneous wound repair in diabetic mice. This study introduces a simple strategy for engineering tunable degradation modalities into synthetic biomaterials, overcoming a key barrier to their use as cell delivery vehicles.
Collapse
Affiliation(s)
- Karen E Martin
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael D Hunckler
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Eunice Chee
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jeremy D Caplin
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Graham F Barber
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Pranav P Kalelkar
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Rebecca S Schneider
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
24
|
Gerardo‐Nava JL, Jansen J, Günther D, Klasen L, Thiebes AL, Niessing B, Bergerbit C, Meyer AA, Linkhorst J, Barth M, Akhyari P, Stingl J, Nagel S, Stiehl T, Lampert A, Leube R, Wessling M, Santoro F, Ingebrandt S, Jockenhoevel S, Herrmann A, Fischer H, Wagner W, Schmitt RH, Kiessling F, Kramann R, De Laporte L. Transformative Materials to Create 3D Functional Human Tissue Models In Vitro in a Reproducible Manner. Adv Healthc Mater 2023; 12:e2301030. [PMID: 37311209 PMCID: PMC11468549 DOI: 10.1002/adhm.202301030] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/21/2023] [Indexed: 06/15/2023]
Abstract
Recreating human tissues and organs in the petri dish to establish models as tools in biomedical sciences has gained momentum. These models can provide insight into mechanisms of human physiology, disease onset, and progression, and improve drug target validation, as well as the development of new medical therapeutics. Transformative materials play an important role in this evolution, as they can be programmed to direct cell behavior and fate by controlling the activity of bioactive molecules and material properties. Using nature as an inspiration, scientists are creating materials that incorporate specific biological processes observed during human organogenesis and tissue regeneration. This article presents the reader with state-of-the-art developments in the field of in vitro tissue engineering and the challenges related to the design, production, and translation of these transformative materials. Advances regarding (stem) cell sources, expansion, and differentiation, and how novel responsive materials, automated and large-scale fabrication processes, culture conditions, in situ monitoring systems, and computer simulations are required to create functional human tissue models that are relevant and efficient for drug discovery, are described. This paper illustrates how these different technologies need to converge to generate in vitro life-like human tissue models that provide a platform to answer health-based scientific questions.
Collapse
|
25
|
Lee M, Abousaud A, Harkins RA, Marin E, Balasubramani D, Churnetski MC, Peker D, Singh A, Koff JL. Important Considerations in the Diagnosis and Management of Post-transplant Lymphoproliferative Disorder. Curr Oncol Rep 2023; 25:883-895. [PMID: 37162742 PMCID: PMC10390257 DOI: 10.1007/s11912-023-01418-0] [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] [Accepted: 03/20/2023] [Indexed: 05/11/2023]
Abstract
PURPOSE OF REVIEW A relative lack of molecular and clinical studies compared to other lymphoid cancers has historically made it difficult to determine optimal management approaches in post-transplant lymphoproliferative disorder (PTLD). We sought to better define the "state of the science" in PTLD by examining recent advances in risk assessment, genomic profiling, and trials of PTLD-directed therapy. RECENT FINDINGS Several major clinical trials highlight risk-stratified sequential therapy incorporating rituximab with or without chemotherapy as a rational treatment strategy in patients with CD20+ PTLD who do not respond to reduction of immunosuppression alone. Epstein Barr virus (EBV)-targeted cytotoxic lymphocytes are a promising approach in patients with relapsed/refractory EBV+ PTLD, but dedicated clinical trials should determine how autologous chimeric antigen receptor T cell therapy (CAR-T) may be safely administered to PTLD patients. Sequencing studies underscore the important effect of EBV infection on PTLD pathogenesis, but comprehensive genomic and tumor microenvironment profiling are needed to identify biomarkers that predict response to treatment in this clinically heterogeneous disease.
Collapse
Affiliation(s)
| | - Aseala Abousaud
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | | | - Ellen Marin
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | | | - Michael C Churnetski
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Deniz Peker
- Department of Pathology, Emory University, Atlanta, GA, USA
| | - Ankur Singh
- Georgia Institute of Technology, Atlanta, GA, USA
| | - Jean L Koff
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
26
|
Affiliation(s)
- Akhilesh K Gaharwar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA.
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, USA.
| | - Irtisha Singh
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA.
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, USA.
- Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, Bryan, TX, USA.
| |
Collapse
|