1
|
Shama KA, Greenberg ZF, Tammame C, He M, Taylor BL. Diseased Tendon Models Demonstrate Influence of Extracellular Matrix Alterations on Extracellular Vesicle Profile. Bioengineering (Basel) 2024; 11:1019. [PMID: 39451395 PMCID: PMC11505312 DOI: 10.3390/bioengineering11101019] [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: 08/16/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Tendons enable movement through their highly aligned extracellular matrix (ECM), predominantly composed of collagen I. Tendinopathies disrupt the structural integrity of tendons by causing fragmentation of collagen fibers, disorganization of fiber bundles, and an increase in glycosaminoglycans and microvasculature, thereby driving the apparent biomechanical and regenerative capacity in patients. Moreover, the complex cellular communication within the tendon microenvironment ultimately dictates the fate between healthy and diseased tendon, wherein extracellular vesicles (EVs) may facilitate the tendon's fate by transporting biomolecules within the tissue. In this study, we aimed to elucidate how the EV functionality is altered in the context of tendon microenvironments by using polycaprolactone (PCL) electrospun scaffolds mimicking healthy and pathological tendon matrices. Scaffolds were characterized for fiber alignment, mechanical properties, and cellular activity. EVs were isolated and analyzed for concentration, heterogeneity, and protein content. Our results show that our mimicked healthy tendon led to an increase in EV secretion and baseline metabolic activity over the mimicked diseased tendon, where reduced EV secretion and a significant increase in metabolic activity over 5 days were observed. These findings suggest that scaffold mechanics may influence EV functionality, offering insights into tendon homeostasis. Future research should further investigate how EV cargo affects the tendon's microenvironment.
Collapse
Affiliation(s)
- Kariman A. Shama
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; (K.A.S.); (C.T.); (M.H.)
| | | | - Chadine Tammame
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; (K.A.S.); (C.T.); (M.H.)
| | - Mei He
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; (K.A.S.); (C.T.); (M.H.)
- Department of Pharmaceutics, University of Florida, Gainesville, FL 32603, USA;
| | - Brittany L. Taylor
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; (K.A.S.); (C.T.); (M.H.)
| |
Collapse
|
2
|
Petrescu DI, Yustein JT, Dasgupta A. Preclinical models for the study of pediatric solid tumors: focus on bone sarcomas. Front Oncol 2024; 14:1388484. [PMID: 39091911 PMCID: PMC11291195 DOI: 10.3389/fonc.2024.1388484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Sarcomas comprise between 10-15% of all pediatric malignancies. Osteosarcoma and Ewing sarcoma are the two most common pediatric bone tumors diagnosed in children and young adults. These tumors are commonly treated with surgery and/or radiation therapy and combination chemotherapy. However, there is a strong need for the development and utilization of targeted therapeutic methods to improve patient outcomes. Towards accomplishing this goal, pre-clinical models for these unique malignancies are of particular importance to design and test experimental therapeutic strategies prior to being introduced to patients due to their origination site and propensity to metastasize. Pre-clinical models offer several advantages for the study of pediatric sarcomas with unique benefits and shortcomings dependent on the type of model. This review addresses the types of pre-clinical models available for the study of pediatric solid tumors, with special attention to the bone sarcomas osteosarcoma and Ewing sarcoma.
Collapse
Affiliation(s)
- D. Isabel Petrescu
- Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA, United States
| | - Jason T. Yustein
- Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA, United States
| | - Atreyi Dasgupta
- The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, Texas Children’s Cancer and Hematology Centers, Houston, TX, United States
| |
Collapse
|
3
|
Holloway AJ, Saito TB, Naqvi KF, Huante MB, Fan X, Lisinicchia JG, Gelman BB, Endsley JJ, Endsley MA. Inhibition of caspase pathways limits CD4 + T cell loss and restores host anti-retroviral function in HIV-1 infected humanized mice with augmented lymphoid tissue. Retrovirology 2024; 21:8. [PMID: 38693565 PMCID: PMC11064318 DOI: 10.1186/s12977-024-00641-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/05/2024] [Indexed: 05/03/2024] Open
Abstract
The study of HIV infection and pathogenicity in physical reservoirs requires a biologically relevant model. The human immune system (HIS) mouse is an established model of HIV infection, but defects in immune tissue reconstitution remain a challenge for examining pathology in tissues. We utilized exogenous injection of the human recombinant FMS-like tyrosine kinase 3 ligand (rFLT-3 L) into the hematopoietic stem cell (HSC) cord blood HIS mouse model to significantly expand the total area of lymph node (LN) and the number of circulating human T cells. The results enabled visualization and quantification of HIV infectivity, CD4 T cell depletion and other measures of pathogenesis in the secondary lymphoid tissues of the spleen and LN. Treatment with the Caspase-1/4 inhibitor VX-765 limited CD4+ T cell loss in the spleen and reduced viral load in both the spleen and axillary LN. In situ hybridization further demonstrated a decrease in viral RNA in both the spleen and LN. Transcriptomic analysis revealed that in vivo inhibition of caspase-1/4 led to an upregulation in host HIV restriction factors including SAMHD1 and APOBEC3A. These findings highlight the use of rFLT-3 L to augment human immune system characteristics in HIS mice to support investigations of HIV pathogenesis and test host directed therapies, though further refinements are needed to further augment LN architecture and cellular populations. The results further provide in vivo evidence of the potential to target inflammasome pathways as an avenue of host-directed therapy to limit immune dysfunction and virus replication in tissue compartments of HIV+ persons.
Collapse
Affiliation(s)
- Alex J Holloway
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555, Galveston, TX, USA
| | - Tais B Saito
- Department of Pathology, University of Texas Medical Branch, 77555, Galveston, TX, USA
- Current at the Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 59840, Hamilton, MT, USA
| | - Kubra F Naqvi
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555, Galveston, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 75390, Dallas, TX, USA
| | - Matthew B Huante
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555, Galveston, TX, USA
| | - Xiuzhen Fan
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555, Galveston, TX, USA
- Department of Medicine, University of Toledo, 43614, Toledo, OH, USA
| | - Joshua G Lisinicchia
- Department of Pathology, University of Texas Medical Branch, 77555, Galveston, TX, USA
| | - Benjamin B Gelman
- Department of Pathology, University of Texas Medical Branch, 77555, Galveston, TX, USA
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555, Galveston, TX, USA
| | - Mark A Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555, Galveston, TX, USA.
| |
Collapse
|
4
|
Luo Y, de Gruijl FR, Vermeer MH, Tensen CP. "Next top" mouse models advancing CTCL research. Front Cell Dev Biol 2024; 12:1372881. [PMID: 38665428 PMCID: PMC11044687 DOI: 10.3389/fcell.2024.1372881] [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: 01/18/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
This review systematically describes the application of in vivo mouse models in studying cutaneous T-cell lymphoma (CTCL), a complex hematological neoplasm. It highlights the diverse research approaches essential for understanding CTCL's intricate pathogenesis and evaluating potential treatments. The review categorizes various mouse models, including xenograft, syngeneic transplantation, and genetically engineered mouse models (GEMMs), emphasizing their contributions to understanding tumor-host interactions, gene functions, and studies on drug efficacy in CTCL. It acknowledges the limitations of these models, particularly in fully replicating human immune responses and early stages of CTCL. The review also highlights novel developments focusing on the potential of skin-targeted GEMMs in studying natural skin lymphoma progression and interactions with the immune system from onset. In conclusion, a balanced understanding of these models' strengths and weaknesses are essential for accelerating the deciphering of CTCL pathogenesis and developing treatment methods. The GEMMs engineered to target specifically skin-homing CD4+ T cells can be the next top mouse models that pave the way for exploring the effects of CTCL-related genes.
Collapse
Affiliation(s)
| | | | | | - Cornelis P. Tensen
- Department of Dermatology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
5
|
Palomares Cabeza V, Fahy N, Kiernan CH, Lolli A, Witte-Bouma J, Fahmy Garcia S, Merino A, Kops N, Ridwan Y, Wolvius EB, Brama PAJ, Hoogduijn MJ, Farrell E. Bone formation by human paediatric marrow stromal cells in a functional allogeneic immune system. Biomaterials 2024; 306:122471. [PMID: 38377846 DOI: 10.1016/j.biomaterials.2024.122471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 02/22/2024]
Abstract
Allogeneic stem-cell based regenerative medicine is a promising approach for bone defect repair. The use of chondrogenically differentiated human marrow stromal cells (MSCs) has been shown to lead to bone formation by endochondral ossification in immunodeficient pre-clinical models. However, an insight into the interactions between the allogeneic immune system and the human MSC-derived bone grafts has not been fully achieved yet. The choice of a potent source of MSCs isolated from pediatric donors with consistent differentiation and high proliferation abilities, as well as low immunogenicity, could increase the chance of success for bone allografts. In this study, we employed an immunodeficient animal model humanised with allogeneic immune cells to study the immune responses towards chondrogenically differentiated human pediatric MSCs (ch-pMSCs). We show that ch-differentiated pMSCs remained non-immunogenic to allogeneic CD4 and CD8 T cells in an in vitro co-culture model. After subcutaneous implantation in mice, ch-pMSC-derived grafts were able to initiate bone mineralisation in the presence of an allogeneic immune system for 3 weeks without the onset of immune responses. Re-exposing the splenocytes of the humanised animals to pMSCs did not trigger further T cell proliferation, suggesting an absence of secondary immune responses. Moreover, ch-pMSCs generated mature bone after 8 weeks of implantation that persisted for up to 6 more weeks in the presence of an allogeneic immune system. These data collectively show that human allogeneic chondrogenically differentiated pediatric MSCs might be a safe and potent option for bone defect repair in the tissue engineering and regenerative medicine setting.
Collapse
Affiliation(s)
- Virginia Palomares Cabeza
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Niamh Fahy
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Applied Science, Technological University of the Shannon: Midlands Midwest, Limerick, Ireland
| | - Caoimhe H Kiernan
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Andrea Lolli
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Shorouk Fahmy Garcia
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Ana Merino
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Nicole Kops
- Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Yanto Ridwan
- AMIE Core Facility, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Eppo B Wolvius
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Pieter A J Brama
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Martin J Hoogduijn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
6
|
Chou SC, Yen CT, Yang YL, Chen SH, Wang JD, Fan MN, Chen LF, Yu IS, Tsai DY, Lin KI, Tao MH, Wu JC, Lin SW. Recapitulating the immune system of hemophilia A patients with inhibitors using immunodeficient mice. Thromb Res 2024; 235:155-163. [PMID: 38341989 DOI: 10.1016/j.thromres.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 12/07/2023] [Accepted: 01/23/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND AND AIM Treating hemophilia A patients who develop inhibitors remains a clinical challenge. A mouse model of hemophilia A can be used to test the efficacy of strategies for inhibitor suppression, but the differences in the immune systems of mice and humans limit its utility. To address this shortcoming, we established a humanized NOD/SCID-IL2rγnull hemophilia A (hu-NSG-HA) mouse model with a severely deficient mouse immune system presenting a patient's adapted immune cells. METHODS AND RESULTS Through intrasplenic injection with patient inhibitor-positive peripheral blood mononuclear cells (PBMCs), utilizing an adeno-associated viral delivery system expressing human BLyS, and regular FVIII challenge, human C19+ B cells were expanded in vivo to secrete anti-FVIII antibodies. Both the inhibitor and the human anti-FVIII IgG, including the predominant subclasses (IgG1 and IgG4) present in the majority of inhibitor patients, were detected in the mouse model. We further segregated and expanded the different clones of human anti-FVIII-secreting cells through subsequent transplantation of splenocytes derived from hu-NSG-HA mice into another NSG-HA mouse. By transplanting a patient's PBMCs into the NSG-HA mouse model, we demonstrated the success of reintroducing a strong anti-FVIII immune response for a short period in mice with the immune systems of inhibitor-positive patients. CONCLUSION Our results demonstrate a potential tool for directly obtaining functional human-derived antigen-specific antibodies and antibody-secreting cells, which may have therapeutic value for testing patient-specific immune responses to treatment options to assist in clinical decisions.
Collapse
Affiliation(s)
- Sheng-Chieh Chou
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Tzu Yen
- Department of Clinical Laboratory Science and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yung-Li Yang
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Huey Chen
- Department of Pediatrics, Shuang Ho Hospital, Ministry of Health and Welfare, Taipei Medical University, New Taipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jiaan-Der Wang
- Children's Medical Center, Taichung Veterans General Hospital, Taiwan; Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung City 407, Taiwan
| | - Meng-Ni Fan
- Department of Clinical Laboratory Science and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Li-Fu Chen
- Department of Clinical Laboratory Science and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - I-Shing Yu
- Laboratory Animal Center, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Dong-Yan Tsai
- Genomics Research Center, Academia Sinica, Taipei, Taiwan; Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Kuo-I Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan; Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Mi-Hua Tao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jui-Ching Wu
- Department of Clinical Laboratory Science and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Shu-Wha Lin
- Department of Clinical Laboratory Science and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
7
|
Du EJ, Muench MO. A Monocytic Barrier to the Humanization of Immunodeficient Mice. Curr Stem Cell Res Ther 2024; 19:959-980. [PMID: 37859310 PMCID: PMC10997744 DOI: 10.2174/011574888x263597231001164351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023]
Abstract
Mice with severe immunodeficiencies have become very important tools for studying foreign cells in an in vivo environment. Xenotransplants can be used to model cells from many species, although most often, mice are humanized through the transplantation of human cells or tissues to meet the needs of medical research. The development of immunodeficient mice is reviewed leading up to the current state-of-the-art strains, such as the NOD-scid-gamma (NSG) mouse. NSG mice are excellent hosts for human hematopoietic stem cell transplants or immune reconstitution through transfusion of human peripheral blood mononuclear cells. However, barriers to full hematopoietic engraftment still remain; notably, the survival of human cells in the circulation is brief, which limits overall hematological and immune reconstitution. Reports have indicated a critical role for monocytic cells - monocytes, macrophages, and dendritic cells - in the clearance of xenogeneic cells from circulation. Various aspects of the NOD genetic background that affect monocytic cell growth, maturation, and function that are favorable to human cell transplantation are discussed. Important receptors, such as SIRPα, that form a part of the innate immune system and enable the recognition and phagocytosis of foreign cells by monocytic cells are reviewed. The development of humanized mouse models has taken decades of work in creating more immunodeficient mice, genetic modification of these mice to express human genes, and refinement of transplant techniques to optimize engraftment. Future advances may focus on the monocytic cells of the host to find ways for further engraftment and survival of xenogeneic cells.
Collapse
Affiliation(s)
- Emily J. Du
- Vitalant Research Institute, 360 Spear Street, Suite 200, San Francisco, CA, 94105, USA
| | - Marcus O. Muench
- Vitalant Research Institute, 360 Spear Street, Suite 200, San Francisco, CA, 94105, USA
- Department of Laboratory Medicine, University of California, San Francisco, CA, 94143, USA
| |
Collapse
|
8
|
Kwee BJ, Li X, Nguyen XX, Campagna C, Lam J, Sung KE. Modeling immunity in microphysiological systems. Exp Biol Med (Maywood) 2023; 248:2001-2019. [PMID: 38166397 PMCID: PMC10800123 DOI: 10.1177/15353702231215897] [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: 01/04/2024] Open
Abstract
There is a need for better predictive models of the human immune system to evaluate safety and efficacy of immunomodulatory drugs and biologics for successful product development and regulatory approvals. Current in vitro models, which are often tested in two-dimensional (2D) tissue culture polystyrene, and preclinical animal models fail to fully recapitulate the function and physiology of the human immune system. Microphysiological systems (MPSs) that can model key microenvironment cues of the human immune system, as well as of specific organs and tissues, may be able to recapitulate specific features of the in vivo inflammatory response. This minireview provides an overview of MPS for modeling lymphatic tissues, immunity at tissue interfaces, inflammatory diseases, and the inflammatory tumor microenvironment in vitro and ex vivo. Broadly, these systems have utility in modeling how certain immunotherapies function in vivo, how dysfunctional immune responses can propagate diseases, and how our immune system can combat pathogens.
Collapse
Affiliation(s)
- Brian J Kwee
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19711, USA
| | - Xiaoqing Li
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Xinh-Xinh Nguyen
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Courtney Campagna
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
| | - Johnny Lam
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Kyung E Sung
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
9
|
Aryee KE, Shultz LD, Burzenski LM, Greiner DL, Brehm MA. NOD-scid IL2rγnull mice lacking TLR4 support human immune system development and the study of human-specific innate immunity. J Leukoc Biol 2023; 113:418-433. [PMID: 36801998 DOI: 10.1093/jleuko/qiac020] [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: 11/15/2021] [Indexed: 01/12/2023] Open
Abstract
Agents that induce inflammation have been used since the 18th century for the treatment of cancer. The inflammation induced by agents such as Toll-like receptor agonists is thought to stimulate tumor-specific immunity in patients and augment control of tumor burden. While NOD-scid IL2rγnull mice lack murine adaptive immunity (T cells and B cells), these mice maintain a residual murine innate immune system that responds to Toll-like receptor agonists. Here we describe a novel NOD-scid IL2rγnull mouse lacking murine TLR4 that fails to respond to lipopolysaccharide. NSG-Tlr4null mice support human immune system engraftment and enable the study of human-specific responses to TLR4 agonists in the absence of the confounding effects of a murine response. Our data demonstrate that specific stimulation of TLR4 activates human innate immune systems and delays the growth kinetics of a human patient-derived xenograft melanoma tumor.
Collapse
Affiliation(s)
- Ken-Edwin Aryee
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, 368 Plantation Street, AS7-2053, Worcester, MA 01605, United States
| | - Leonard D Shultz
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME 04609, United States
| | - Lisa M Burzenski
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME 04609, United States
| | - Dale L Greiner
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, 368 Plantation Street, AS7-2053, Worcester, MA 01605, United States
| | - Michael A Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, 368 Plantation Street, AS7-2053, Worcester, MA 01605, United States
| |
Collapse
|
10
|
Chuprin J, Buettner H, Seedhom MO, Greiner DL, Keck JG, Ishikawa F, Shultz LD, Brehm MA. Humanized mouse models for immuno-oncology research. Nat Rev Clin Oncol 2023; 20:192-206. [PMID: 36635480 PMCID: PMC10593256 DOI: 10.1038/s41571-022-00721-2] [Citation(s) in RCA: 96] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/14/2023]
Abstract
Immunotherapy has emerged as a promising treatment paradigm for many malignancies and is transforming the drug development landscape. Although immunotherapeutic agents have demonstrated clinical efficacy, they are associated with variable clinical responses, and substantial gaps remain in our understanding of their mechanisms of action and specific biomarkers of response. Currently, the number of preclinical models that faithfully recapitulate interactions between the human immune system and tumours and enable evaluation of human-specific immunotherapies in vivo is limited. Humanized mice, a term that refers to immunodeficient mice co-engrafted with human tumours and immune components, provide several advantages for immuno-oncology research. In this Review, we discuss the benefits and challenges of the currently available humanized mice, including specific interactions between engrafted human tumours and immune components, the development and survival of human innate immune populations in these mice, and approaches to study mice engrafted with matched patient tumours and immune cells. We highlight the latest advances in the generation of humanized mouse models, with the aim of providing a guide for their application to immuno-oncology studies with potential for clinical translation.
Collapse
Affiliation(s)
- Jane Chuprin
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Molecular, Cell and Cancer Biology, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Hannah Buettner
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Surgery, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Mina O Seedhom
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dale L Greiner
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | | | | | - Michael A Brehm
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
11
|
Brunetti JE, Kitsera M, Muñoz-Fontela C, Rodríguez E. Use of Hu-PBL Mice to Study Pathogenesis of Human-Restricted Viruses. Viruses 2023; 15:228. [PMID: 36680271 PMCID: PMC9866769 DOI: 10.3390/v15010228] [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: 12/22/2022] [Revised: 01/07/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Different humanized mouse models have been developed to study human diseases such as autoimmune illnesses, cancer and viral infections. These models are based on the use of immunodeficient mouse strains that are transplanted with human tissues or human immune cells. Among the latter, mice transplanted with hematopoietic stem cells have been widely used to study human infectious diseases. However, mouse models built upon the transplantation of donor-specific mature immune cells are still under development, especially in the field of viral infections. These models can retain the unique immune memory of the donor, making them suitable for the study of correlates of protection upon natural infection or vaccination. Here, we will review some of these models and how they have been applied to virology research. Moreover, the future applications and the potential of these models to design therapies against human viral infections are discussed.
Collapse
Affiliation(s)
| | - Maksym Kitsera
- Bernhard-Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - César Muñoz-Fontela
- Bernhard-Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Borstel-Lübeck, 38124 Braunschweig, Germany
| | - Estefanía Rodríguez
- Bernhard-Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Borstel-Lübeck, 38124 Braunschweig, Germany
| |
Collapse
|
12
|
Ashida Y, Himori K, Tokuda N, Naito A, Yamauchi N, Takenaka-Ninagawa N, Aoki Y, Sakurai H, Yamada T. Dissociation of SH3 and cysteine rich domain 3 and junctophilin 1 from dihydropyridine receptor in dystrophin-deficient muscles. Am J Physiol Cell Physiol 2022; 323:C885-C895. [PMID: 35912995 DOI: 10.1152/ajpcell.00163.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The disruption of excitation-contraction (EC) coupling and subsequent reduction in Ca2+ release from the sarcoplasmic reticulum (SR) have been shown to account for muscle weakness seen in patients with Duchenne muscular dystrophy (DMD). Here, we examined the mechanisms underlying EC uncoupling in skeletal muscles from mdx52 and DMD-null/NSG mice, animal models for DMD, focusing on the SH3 and cysteine rich domain 3 (STAC3) and junctophilin 1 (JP1), which link the dihydropyridine receptor (DHPR) in the transverse tubule and the ryanodine receptor 1 in the SR. The isometric plantarflexion torque normalized to muscle weight of whole plantar flexor muscles was depressed in mdx52 and DMD-null/NSG mice compared to their control mice. This was accompanied by increased autolysis of calpain-1, decreased levels of STAC3 and JP1 content, and dissociation of STAC3 and JP1 from DHPR-α1s in gastrocnemius muscles. Moreover, in vitro mechanistic experiments demonstrated that STAC3 and JP1 underwent Ca2+-dependent proteolysis which was less pronounced in dystrophin-deficient muscles where calpastatin, the endogenous calpain inhibitor, was upregulated. Eccentric contractions further enhanced autolysis of calpain-1 and proteolysis of STAC3 and JP1 that were associated with severe torque depression in gastrocnemius muscles from DMD-null/NSG mice. These data suggest that Ca2+-dependent proteolysis of STAC3 and JP1 may be an essential factor causing muscle weakness due to EC coupling failure in dystrophin-deficient muscles.
Collapse
Affiliation(s)
- Yuki Ashida
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Koichi Himori
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Nao Tokuda
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Azuma Naito
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Nao Yamauchi
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | | | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Takashi Yamada
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
13
|
Gomez-Florit M, Labrador-Rached CJ, Domingues RM, Gomes ME. The tendon microenvironment: Engineered in vitro models to study cellular crosstalk. Adv Drug Deliv Rev 2022; 185:114299. [PMID: 35436570 DOI: 10.1016/j.addr.2022.114299] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022]
Abstract
Tendinopathy is a multi-faceted pathology characterized by alterations in tendon microstructure, cellularity and collagen composition. Challenged by the possibility of regenerating pathological or ruptured tendons, the healing mechanisms of this tissue have been widely researched over the past decades. However, so far, most of the cellular players and processes influencing tendon repair remain unknown, which emphasizes the need for developing relevant in vitro models enabling to study the complex multicellular crosstalk occurring in tendon microenvironments. In this review, we critically discuss the insights on the interaction between tenocytes and the other tendon resident cells that have been devised through different types of existing in vitro models. Building on the generated knowledge, we stress the need for advanced models able to mimic the hierarchical architecture, cellularity and physiological signaling of tendon niche under dynamic culture conditions, along with the recreation of the integrated gradients of its tissue interfaces. In a forward-looking vision of the field, we discuss how the convergence of multiple bioengineering technologies can be leveraged as potential platforms to develop the next generation of relevant in vitro models that can contribute for a deeper fundamental knowledge to develop more effective treatments.
Collapse
|
14
|
Calejo I, Labrador‐Rached CJ, Gomez‐Florit M, Docheva D, Reis RL, Domingues RMA, Gomes ME. Bioengineered 3D Living Fibers as In Vitro Human Tissue Models of Tendon Physiology and Pathology. Adv Healthc Mater 2022; 11:e2102863. [PMID: 35596614 DOI: 10.1002/adhm.202102863] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/07/2022] [Indexed: 12/12/2022]
Abstract
Clinically relevant in vitro models of human tissue's health and disease are urgently needed for a better understanding of biological mechanisms essential for the development of novel therapies. Herein, physiological (healthy) and pathological (disease) tendon states are bioengineered by coupling the biological signaling of platelet lysate components with controlled 3D architectures of electrospun microfibers to drive the fate of human tendon cells in different composite living fibers (CLFs). In the CLFs-healthy model, tendon cells adopt a high cytoskeleton alignment and elongation, express tendon-related markers (scleraxis, tenomodulin, and mohawk) and deposit a dense tenogenic matrix. In contrast, cell crowding with low preferential orientation, high matrix deposition, and phenotypic drift leading to increased expression of nontendon related and fibrotic markers, are characteristics of the CLFs-diseased model. This diseased-like profile, also reflected in the increase of COL3/COL1 ratio, is further evident by the imbalance between matrix remodeling and degradation effectors, characteristic of tendinopathy. In summary, microengineered 3D in vitro models of human tendon healthy and diseased states are successfully fabricated. Most importantly, these innovative and versatile microphysiological models offer major advantages over currently used systems, holding promise for drugs screening and development of new therapies.
Collapse
Affiliation(s)
- Isabel Calejo
- 3B's Research Group i3Bs—Research Institute on Biomaterials Biodegradables and Biomimetics University of Minho 4805‐017 Barco Guimarães Portugal
| | - Claudia J. Labrador‐Rached
- 3B's Research Group i3Bs—Research Institute on Biomaterials Biodegradables and Biomimetics University of Minho 4805‐017 Barco Guimarães Portugal
| | - Manuel Gomez‐Florit
- 3B's Research Group i3Bs—Research Institute on Biomaterials Biodegradables and Biomimetics University of Minho 4805‐017 Barco Guimarães Portugal
| | - Denitsa Docheva
- Experimental Trauma Surgery Department of Trauma Surgery University Hospital Regensburg Franz‐Josef Strauss‐Allee 11 93053 Regensburg Germany
| | - Rui L. Reis
- 3B's Research Group i3Bs—Research Institute on Biomaterials Biodegradables and Biomimetics University of Minho 4805‐017 Barco Guimarães Portugal
| | - Rui M. A. Domingues
- 3B's Research Group i3Bs—Research Institute on Biomaterials Biodegradables and Biomimetics University of Minho 4805‐017 Barco Guimarães Portugal
| | - Manuela E. Gomes
- 3B's Research Group i3Bs—Research Institute on Biomaterials Biodegradables and Biomimetics University of Minho 4805‐017 Barco Guimarães Portugal
| |
Collapse
|
15
|
Nauman G, Danzl NM, Lee J, Borsotti C, Madley R, Fu J, Hölzl MA, Dahmani A, Dorronsoro Gonzalez A, Chavez É, Campbell SR, Yang S, Satwani P, Liu K, Sykes M. Defects in Long-Term APC Repopulation Ability of Adult Human Bone Marrow Hematopoietic Stem Cells (HSCs) Compared with Fetal Liver HSCs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1652-1663. [PMID: 35315788 PMCID: PMC8976823 DOI: 10.4049/jimmunol.2100966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/25/2022] [Indexed: 04/28/2023]
Abstract
Immunodeficient mice reconstituted with immune systems from patients, or personalized immune (PI) mice, are powerful tools for understanding human disease. Compared with immunodeficient mice transplanted with human fetal thymus tissue and fetal liver-derived CD34+ cells administered i.v. (Hu/Hu mice), PI mice, which are transplanted with human fetal thymus and adult bone marrow (aBM) CD34+ cells, demonstrate reduced levels of human reconstitution. We characterized APC and APC progenitor repopulation in human immune system mice and detected significant reductions in blood, bone marrow (BM), and splenic APC populations in PI compared with Hu/Hu mice. APC progenitors and hematopoietic stem cells (HSCs) were less abundant in aBM CD34+ cells compared with fetal liver-derived CD34+ cell preparations, and this reduction in APC progenitors was reflected in the BM of PI compared with Hu/Hu mice 14-20 wk posttransplant. The number of HSCs increased in PI mice compared with the originally infused BM cells and maintained functional repopulation potential, because BM from some PI mice 28 wk posttransplant generated human myeloid and lymphoid cells in secondary recipients. Moreover, long-term PI mouse BM contained functional T cell progenitors, evidenced by thymopoiesis in thymic organ cultures. Injection of aBM cells directly into the BM cavity, transgenic expression of hematopoietic cytokines, and coinfusion of human BM-derived mesenchymal stem cells synergized to enhance long-term B cell and monocyte levels in PI mice. These improvements allow a sustained time frame of 18-22 wk where APCs and T cells are present and greater flexibility for modeling immune disease pathogenesis and immunotherapies in PI mice.
Collapse
Affiliation(s)
- Grace Nauman
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Nichole M Danzl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Jaeyop Lee
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Chiara Borsotti
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Rachel Madley
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Jianing Fu
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Markus A Hölzl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Alexander Dahmani
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Akaitz Dorronsoro Gonzalez
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Éstefania Chavez
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Sean R Campbell
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Suxiao Yang
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Prakash Satwani
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Pediatrics, Columbia University Medical Center, Columbia University, New York, NY
| | - Kang Liu
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT; and
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY;
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Surgery, Columbia University Medical Center, Columbia University, New York, NY
| |
Collapse
|
16
|
Mesenchymal stromal cell apoptosis is required for their therapeutic function. Nat Commun 2021; 12:6495. [PMID: 34764248 PMCID: PMC8586224 DOI: 10.1038/s41467-021-26834-3] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 10/21/2021] [Indexed: 12/21/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) ameliorate a wide range of diseases in preclinical models, but the lack of clarity around their mechanisms of action has impeded their clinical utility. The therapeutic effects of MSCs are often attributed to bioactive molecules secreted by viable MSCs. However, we found that MSCs underwent apoptosis in the lung after intravenous administration, even in the absence of host cytotoxic or alloreactive cells. Deletion of the apoptotic effectors BAK and BAX prevented MSC death and attenuated their immunosuppressive effects in disease models used to define MSC potency. Mechanistically, apoptosis of MSCs and their efferocytosis induced changes in metabolic and inflammatory pathways in alveolar macrophages to effect immunosuppression and reduce disease severity. Our data reveal a mode of action whereby the host response to dying MSCs is key to their therapeutic effects; findings that have broad implications for the effective translation of cell-based therapies. Mesenchymal stromal cells (MSCs) demonstrate therapeutic benefits in multiple diseases, but the mechanisms remain unclear as infused MSCs do not persist in the body. Here, the authors show that MSC apoptosis is an important mechanistic element, as MSCs rendered genetically incapable of apoptosis lose their ability to ameliorate disease.
Collapse
|
17
|
Genome editing in large animal models. Mol Ther 2021; 29:3140-3152. [PMID: 34601132 DOI: 10.1016/j.ymthe.2021.09.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/26/2021] [Accepted: 09/26/2021] [Indexed: 12/21/2022] Open
Abstract
Although genome editing technologies have the potential to revolutionize the way we treat human diseases, barriers to successful clinical implementation remain. Increasingly, preclinical large animal models are being used to overcome these barriers. In particular, the immunogenicity and long-term safety of novel gene editing therapeutics must be evaluated rigorously. However, short-lived small animal models, such as mice and rats, cannot address secondary pathologies that may arise years after a gene editing treatment. Likewise, immunodeficient mouse models by definition lack the ability to quantify the host immune response to a novel transgene or gene-edited locus. Large animal models, including dogs, pigs, and non-human primates (NHPs), bear greater resemblance to human anatomy, immunology, and lifespan and can be studied over longer timescales with clinical dosing regimens that are more relevant to humans. These models allow for larger scale and repeated blood and tissue sampling, enabling greater depth of study and focus on rare cellular subsets. Here, we review current progress in the development and evaluation of novel genome editing therapies in large animal models, focusing on applications in human immunodeficiency virus 1 (HIV-1) infection, cancer, and genetic diseases including hemoglobinopathies, Duchenne muscular dystrophy (DMD), hypercholesterolemia, and inherited retinal diseases.
Collapse
|
18
|
Emmanuel T, Mistegård J, Bregnhøj A, Johansen C, Iversen L. Tissue-Resident Memory T Cells in Skin Diseases: A Systematic Review. Int J Mol Sci 2021; 22:ijms22169004. [PMID: 34445713 PMCID: PMC8396505 DOI: 10.3390/ijms22169004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/09/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023] Open
Abstract
In health, the non-recirculating nature and long-term persistence of tissue-resident memory T cells (TRMs) in tissues protects against invading pathogens. In disease, pathogenic TRMs contribute to the recurring traits of many skin diseases. We aimed to conduct a systematic literature review on the current understanding of the role of TRMs in skin diseases and identify gaps as well as future research paths. EMBASE, PubMed, SCOPUS, Web of Science, Clinicaltrials.gov and WHO Trials Registry were searched systematically for relevant studies from their inception to October 2020. Included studies were reviewed independently by two authors. This study was conducted in accordance with the PRISMA-S guidelines. This protocol was registered with the PROSPERO database (ref: CRD42020206416). We identified 96 studies meeting the inclusion criteria. TRMs have mostly been investigated in murine skin and in relation to infectious skin diseases. Pathogenic TRMs have been characterized in various skin diseases including psoriasis, vitiligo and cutaneous T-cell lymphoma. Studies are needed to discover biomarkers that may delineate TRMs poised for pathogenic activity in skin diseases and establish to which extent TRMs are contingent on the local skin microenvironment. Additionally, future studies may investigate the effects of current treatments on the persistence of pathogenic TRMs in human skin.
Collapse
|
19
|
Ko Y, Jeong YH, Seo JH, Lee JA. Development of a Bioluminescent Human Osteosarcoma Model in Humanized NSG Mice: A Pilot Study. In Vivo 2021; 35:2151-2157. [PMID: 34182491 DOI: 10.21873/invivo.12485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND/AIM Osteosarcoma is the most common type of bone cancer, but current therapeutic interventions remain largely insufficient. The development of new treatment strategies is needed, and moreover, optimal rodent models are necessary for testing the efficacy of new treatment modalities of osteosarcoma. Humanized mice carry human hematopoietic and immune systems, and are considered an ideal tool to study human diseases including cancer immunology. Herein, we performed a preliminary study toward developing an in vivo bioluminescent osteosarcoma model using humanized immunodeficient (NSG) mice. MATERIALS AND METHODS To establish the xenograft and orthotopic mouse model, NSG mice engrafted with human CD34+ hematopoietic stem cells were injected with luciferase-expressing KHOS/NP cells at two different time points. Bioluminescence images were obtained to monitor in vivo tumor growth and metastasis. Influence of the degree of human cell engraftment on tumor growth and metastatic behavior was analyzed and compared between the two groups. RESULTS KHOS/NP-luc cells injected in humanized NSG mice formed macroscopic tumors. The percentage of human CD45+ cells in these models was similar, but the percentage of human CD45+CD3+ and their subset was higher in the late-injection group compared to that of the early-injection group. The rate of KHOS/NP tumor growth was higher in the early-injection group than in the late-injection group. In the present study, human hematopoietic cell engraftment was not influenced by KHOS/NP cell injection, but KHOS/NP osteosarcoma showed more aggressive behavior in the early-injection group than that in the late-injection group, forming larger tumor volumes and earlier metastases. CONCLUSION The results indicated that tumor growth and progression in humanized NSG mice may have been influenced by higher levels of human cell engraftment, especially T cells. Although there exist some limitations to our study, our preliminary results can provide the basis for the development of a humanized osteosarcoma mouse model.
Collapse
Affiliation(s)
- Yunmi Ko
- Department of Pediatrics, Center for Pediatric Cancer, National Cancer Center, Goyang, Republic of Korea.,Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Yeon Ho Jeong
- Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Jin-Hee Seo
- Laboratory Animal Team, Radiation Medicine Support Center, Division of Fusion Radiology Research, Korea Institute of radiological & Medical Sciences, Seoul, Republic of Korea
| | - Jun Ah Lee
- Department of Pediatrics, Center for Pediatric Cancer, National Cancer Center, Goyang, Republic of Korea;
| |
Collapse
|
20
|
Somogyi E, Csiszovszki Z, Molnár L, Lőrincz O, Tóth J, Pattijn S, Schockaert J, Mazy A, Miklós I, Pántya K, Páles P, Tőke ER. A Peptide Vaccine Candidate Tailored to Individuals' Genetics Mimics the Multi-Targeted T Cell Immunity of COVID-19 Convalescent Subjects. Front Genet 2021; 12:684152. [PMID: 34249101 PMCID: PMC8261158 DOI: 10.3389/fgene.2021.684152] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/24/2021] [Indexed: 01/21/2023] Open
Abstract
Long-term immunity to coronaviruses likely stems from T cell activity. We present here a novel approach for the selection of immunoprevalent SARS-CoV-2-derived T cell epitopes using an in silico cohort of HLA-genotyped individuals with different ethnicities. Nine 30-mer peptides derived from the four major structural proteins of SARS-CoV-2 were selected and included in a peptide vaccine candidate to recapitulate the broad virus-specific T cell responses observed in natural infection. PolyPEPI-SCoV-2-specific, polyfunctional CD8+ and CD4+ T cells were detected in each of the 17 asymptomatic/mild COVID-19 convalescents' blood against on average seven different vaccine peptides. Furthermore, convalescents' complete HLA-genotype predicted their T cell responses to SARS-CoV-2-derived peptides with 84% accuracy. Computational extrapolation of this relationship to a cohort of 16,000 HLA-genotyped individuals with 16 different ethnicities suggest that PolyPEPI-SCoV-2 vaccination will likely elicit multi-antigenic T cell responses in 98% of individuals, independent of ethnicity. PolyPEPI-SCoV-2 administered with Montanide ISA 51 VG generated robust, Th1-biased CD8+, and CD4+ T cell responses against all represented proteins, as well as binding antibodies upon subcutaneous injection into BALB/c and hCD34+ transgenic mice modeling human immune system. These results have implications for the development of global, highly immunogenic, T cell-focused vaccines against various pathogens and diseases.
Collapse
Affiliation(s)
- Eszter Somogyi
- Treos Bio Ltd., London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Zsolt Csiszovszki
- Treos Bio Ltd., London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Levente Molnár
- Treos Bio Ltd., London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Orsolya Lőrincz
- Treos Bio Ltd., London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - József Tóth
- Treos Bio Ltd., London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Sofie Pattijn
- ImmunXperts Société Anonyme, A Nexelis Group Company, Gosselies, Belgium
| | - Jana Schockaert
- ImmunXperts Société Anonyme, A Nexelis Group Company, Gosselies, Belgium
| | - Aurélie Mazy
- ImmunXperts Société Anonyme, A Nexelis Group Company, Gosselies, Belgium
| | - István Miklós
- Treos Bio Ltd., London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
- Alfréd Rényi Institute of Mathematics, Eötvös Loránd Research Network, Budapest, Hungary
| | - Katalin Pántya
- Treos Bio Ltd., London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Péter Páles
- Treos Bio Ltd., London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Enikő R. Tőke
- Treos Bio Ltd., London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| |
Collapse
|
21
|
Pellizzari G, Martinez O, Crescioli S, Page R, Di Meo A, Mele S, Chiaruttini G, Hoinka J, Batruch I, Prassas I, Grandits M, López-Abente J, Bugallo-Blanco E, Ward M, Bax HJ, French E, Cheung A, Lombardi S, Figini M, Lacy KE, Diamandis EP, Josephs DH, Spicer J, Papa S, Karagiannis SN. Immunotherapy using IgE or CAR T cells for cancers expressing the tumor antigen SLC3A2. J Immunother Cancer 2021; 9:jitc-2020-002140. [PMID: 34112739 PMCID: PMC8194339 DOI: 10.1136/jitc-2020-002140] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2021] [Indexed: 01/21/2023] Open
Abstract
Background Cancer immunotherapy with monoclonal antibodies and chimeric antigen receptor (CAR) T cell therapies can benefit from selection of new targets with high levels of tumor specificity and from early assessments of efficacy and safety to derisk potential therapies. Methods Employing mass spectrometry, bioinformatics, immuno-mass spectrometry and CRISPR/Cas9 we identified the target of the tumor-specific SF-25 antibody. We engineered IgE and CAR T cell immunotherapies derived from the SF-25 clone and evaluated potential for cancer therapy. Results We identified the target of the SF-25 clone as the tumor-associated antigen SLC3A2, a cell surface protein with key roles in cancer metabolism. We generated IgE monoclonal antibody, and CAR T cell immunotherapies each recognizing SLC3A2. In concordance with preclinical and, more recently, clinical findings with the first-in-class IgE antibody MOv18 (recognizing the tumor-associated antigen Folate Receptor alpha), SF-25 IgE potentiated Fc-mediated effector functions against cancer cells in vitro and restricted human tumor xenograft growth in mice engrafted with human effector cells. The antibody did not trigger basophil activation in cancer patient blood ex vivo, suggesting failure to induce type I hypersensitivity, and supporting safe therapeutic administration. SLC3A2-specific CAR T cells demonstrated cytotoxicity against tumor cells, stimulated interferon-γ and interleukin-2 production in vitro. In vivo SLC3A2-specific CAR T cells significantly increased overall survival and reduced growth of subcutaneous PC3-LN3-luciferase xenografts. No weight loss, manifestations of cytokine release syndrome or graft-versus-host disease, were detected. Conclusions These findings identify efficacious and potentially safe tumor-targeting of SLC3A2 with novel immune-activating antibody and genetically modified cell therapies.
Collapse
Affiliation(s)
- Giulia Pellizzari
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK
| | - Olivier Martinez
- Immunoengineering Group, King's College London, London, England, UK
| | - Silvia Crescioli
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK
| | - Robert Page
- Immunoengineering Group, King's College London, London, England, UK
| | - Ashley Di Meo
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Silvia Mele
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK
| | - Giulia Chiaruttini
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK
| | - Jan Hoinka
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Ihor Batruch
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ioannis Prassas
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Melanie Grandits
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK
| | - Jacobo López-Abente
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK
| | | | | | - Heather J Bax
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK
| | - Elise French
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK
| | - Anthony Cheung
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK.,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, London, England, UK
| | - Sara Lombardi
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK.,School of Cancer and Pharmaceutical Sciences, King's College London, London, England, UK
| | - Mariangela Figini
- Biomarker Unit, Dipartimento di Ricerca Applicata e Sviluppo Tecnologico (DRAST), Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Katie E Lacy
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK
| | - Eleftherios P Diamandis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, Canada
| | - Debra H Josephs
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK.,Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, England, UK
| | - James Spicer
- School of Cancer and Pharmaceutical Sciences, King's College London, London, England, UK
| | - Sophie Papa
- Immunoengineering Group, King's College London, London, England, UK .,Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, England, UK
| | - Sophia N Karagiannis
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, England, UK .,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, London, England, UK
| |
Collapse
|
22
|
Jin KT, Du WL, Lan HR, Liu YY, Mao CS, Du JL, Mou XZ. Development of humanized mouse with patient-derived xenografts for cancer immunotherapy studies: A comprehensive review. Cancer Sci 2021; 112:2592-2606. [PMID: 33938090 PMCID: PMC8253285 DOI: 10.1111/cas.14934] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy has revolutionized cancer treatment, however, not all tumor types and patients are completely responsive to this approach. Establishing predictive pre-clinical models would allow for more accurate and practical immunotherapeutic drug development. Mouse models are extensively used as in vivo system for biomedical research. However, due to the significant differences between rodents and human, it is impossible to translate most of the findings from mouse models to human. Pharmacological development and advancing personalized medicine using patient-derived xenografts relies on producing mouse models in which murine cells and genes are substituted with their human equivalent. Humanized mice (HM) provide a suitable platform to evaluate xenograft growth in the context of a human immune system. In this review, we discussed recent advances in the generation and application of HM models. We also reviewed new insights into the basic mechanisms, pre-clinical evaluation of onco-immunotherapies, current limitations in the application of these models as well as available improvement strategies. Finally, we pointed out some issues for future studies.
Collapse
Affiliation(s)
- Ke-Tao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Wen-Lin Du
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Huan-Rong Lan
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Yu-Yao Liu
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Chun-Sen Mao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jin-Lin Du
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xiao-Zhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
23
|
Terahara K, Iwabuchi R, Tsunetsugu-Yokota Y. Perspectives on Non-BLT Humanized Mouse Models for Studying HIV Pathogenesis and Therapy. Viruses 2021; 13:v13050776. [PMID: 33924786 PMCID: PMC8145733 DOI: 10.3390/v13050776] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
A variety of humanized mice, which are reconstituted only with human hematopoietic stem cells (HSC) or with fetal thymus and HSCs, have been developed and widely utilized as in vivo animal models of HIV-1 infection. The models represent some aspects of HIV-mediated pathogenesis in humans and are useful for the evaluation of therapeutic regimens. However, there are several limitations in these models, including their incomplete immune responses and poor distribution of human cells to the secondary lymphoid tissues. These limitations are common in many humanized mouse models and are critical issues that need to be addressed. As distinct defects exist in each model, we need to be cautious about the experimental design and interpretation of the outcomes obtained using humanized mice. Considering this point, we mainly characterize the current conventional humanized mouse reconstituted only with HSCs and describe past achievements in this area, as well as the potential contributions of the humanized mouse models for the study of HIV pathogenesis and therapy. We also discuss the use of various technologies to solve the current problems. Humanized mice will contribute not only to the pre-clinical evaluation of anti-HIV regimens, but also to a deeper understanding of basic aspects of HIV biology.
Collapse
Affiliation(s)
- Kazutaka Terahara
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (K.T.); (R.I.)
| | - Ryutaro Iwabuchi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (K.T.); (R.I.)
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo 162-8480, Japan
| | - Yasuko Tsunetsugu-Yokota
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (K.T.); (R.I.)
- Department of Medical Technology, School of Human Sciences, Tokyo University of Technology, Tokyo 144-8535, Japan
- Correspondence: or ; Tel.: +81-3-6424-2223
| |
Collapse
|
24
|
Unlocking the Non-IgE-Mediated Pseudo-Allergic Reaction Puzzle with Mas-Related G-Protein Coupled Receptor Member X2 (MRGPRX2). Cells 2021; 10:cells10051033. [PMID: 33925682 PMCID: PMC8146469 DOI: 10.3390/cells10051033] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/19/2021] [Accepted: 04/25/2021] [Indexed: 12/17/2022] Open
Abstract
Mas-related G-protein coupled receptor member X2 (MRGPRX2) is a class A GPCR expressed on mast cells. Mast cells are granulated tissue-resident cells known for host cell response, allergic response, and vascular homeostasis. Immunoglobulin E receptor (FcεRI)-mediated mast cell activation is a well-studied and recognized mechanism of allergy and hypersensitivity reactions. However, non-IgE-mediated mast cell activation is less explored and is not well recognized. After decades of uncertainty, MRGPRX2 was discovered as the receptor responsible for non-IgE-mediated mast cells activation. The puzzle of non-IgE-mediated pseudo-allergic reaction is unlocked by MRGPRX2, evidenced by a plethora of reported endogenous and exogenous MRGPRX2 agonists. MRGPRX2 is exclusively expressed on mast cells and exhibits varying affinity for many molecules such as antimicrobial host defense peptides, neuropeptides, and even US Food and Drug Administration-approved drugs. The discovery of MRGPRX2 has changed our understanding of mast cell biology and filled the missing link of the underlying mechanism of drug-induced MC degranulation and pseudo-allergic reactions. These non-canonical characteristics render MRGPRX2 an intriguing player in allergic diseases. In the present article, we reviewed the emerging role of MRGPRX2 as a non-IgE-mediated mechanism of mast cell activation in pseudo-allergic reactions. We have presented an overview of mast cells, their receptors, structural insight into MRGPRX2, MRGPRX2 agonists and antagonists, the crucial role of MRGPRX2 in pseudo-allergic reactions, current challenges, and the future research direction.
Collapse
|
25
|
Nagle VL, Henry KE, Hertz CAJ, Graham MS, Campos C, Parada LF, Pandit-Taskar N, Schietinger A, Mellinghoff IK, Lewis JS. Imaging Tumor-Infiltrating Lymphocytes in Brain Tumors with [ 64Cu]Cu-NOTA-anti-CD8 PET. Clin Cancer Res 2021; 27:1958-1966. [PMID: 33495310 PMCID: PMC8026513 DOI: 10.1158/1078-0432.ccr-20-3243] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/15/2020] [Accepted: 01/15/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Glioblastoma (GBM) is the most common malignant brain tumor in adults. Various immunotherapeutic approaches to improve patient survival are being developed, but the molecular mechanisms of immunotherapy resistance are currently unknown. Here, we explored the ability of a humanized radiolabeled CD8-targeted minibody to noninvasively quantify tumor-infiltrating CD8-positive (CD8+) T cells using PET. EXPERIMENTAL DESIGN We generated a peripheral blood mononuclear cell (PBMC) humanized immune system (HIS) mouse model and quantified the absolute number of CD8+ T cells by flow cytometry relative to the [64Cu]Cu-NOTA-anti-CD8 PET signal. To evaluate a patient-derived orthotopic GBM HIS model, we intracranially injected cells into NOG mice, humanized cohorts with multiple HLA-matched PBMC donors, and quantified CD8+ tumor-infiltrating lymphocytes by IHC. To determine whether [64Cu]Cu-NOTA-anti-CD8 images brain parenchymal T-cell infiltrate in GBM tumors, we performed PET and autoradiography and subsequently stained serial sections of brain tumor tissue by IHC for CD8+ T cells. RESULTS Nontumor-bearing NOG mice injected with human PBMCs showed prominent [64Cu]Cu-NOTA-anti-CD8 uptake in the spleen and minimal radiotracer localization to the normal brain. NOG mice harboring intracranial human GBMs yielded high-resolution PET images of tumor-infiltrating CD8+ T cells. Radiotracer retention correlated with CD8+ T-cell numbers in spleen and tumor tissue. Our study demonstrates the ability of [64Cu]Cu-NOTA-anti-CD8 PET to quantify peripheral and tumor-infiltrating CD8+ T cells in brain tumors. CONCLUSIONS Human CD8+ T cells infiltrate an orthotopic GBM in a donor-dependent manner. Furthermore, [64Cu]Cu-NOTA-anti-CD8 quantitatively images both peripheral and brain parenchymal human CD8+ T cells.
Collapse
Affiliation(s)
- Veronica L Nagle
- Department of Pharmacology, Weill Cornell Medical College, New York, New York
| | - Kelly E Henry
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charli Ann J Hertz
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maya S Graham
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Carl Campos
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Luis F Parada
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrea Schietinger
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ingo K Mellinghoff
- Department of Pharmacology, Weill Cornell Medical College, New York, New York.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason S Lewis
- Department of Pharmacology, Weill Cornell Medical College, New York, New York.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
26
|
Iwabuchi R, Ide K, Terahara K, Wagatsuma R, Iwaki R, Matsunaga H, Tsunetsugu-Yokota Y, Takeyama H, Takahashi Y. Development of an Inflammatory CD14 + Dendritic Cell Subset in Humanized Mice. Front Immunol 2021; 12:643040. [PMID: 33790912 PMCID: PMC8005643 DOI: 10.3389/fimmu.2021.643040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Humanized mouse models are attractive experimental models for analyzing the development and functions of human dendritic cells (DCs) in vivo. Although various types of DC subsets, including DC type 3 (DC3s), have been identified in humans, it remains unclear whether humanized mice can reproduce heterogeneous DC subsets. CD14, classically known as a monocyte/macrophage marker, is reported as an indicator of DC3s. We previously observed that some CD14+ myeloid cells expressed CD1c, a pan marker for bona fide conventional DC2 (cDC2s), in humanized mouse models in which human FLT3L and GM-CSF genes were transiently expressed using in vivo transfection (IVT). Here, we aimed to elucidate the identity of CD14+CD1c+ DC-like cells in humanized mouse models. We found that CD14+CD1c+ cells were phenotypically different from cDC2s; CD14+CD1c+ cells expressed CD163 but not CD5, whereas cDC2s expressed CD5 but not CD163. Furthermore, CD14+CD1c+ cells primed and polarized naïve CD4+ T cells toward IFN-γ+ Th1 cells more profoundly than cDC2s. Transcriptional analysis revealed that CD14+CD1c+ cells expressed several DC3-specific transcripts, such as CD163, S100A8, and S100A9, and were clearly segregated from cDC2s and monocytes. When lipopolysaccharide was administered to the humanized mice, the frequency of CD14+CD1c+ cells producing IL-6 and TNF-α was elevated, indicating a pro-inflammatory signature. Thus, humanized mice are able to sustain development of functional CD14+CD1c+ DCs, which are equivalent to DC3 subset observed in humans, and they could be useful for analyzing the development and function of DC3s in vivo.
Collapse
Affiliation(s)
- Ryutaro Iwabuchi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan.,Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Keigo Ide
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan.,Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Kazutaka Terahara
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ryota Wagatsuma
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - Rieko Iwaki
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hiroko Matsunaga
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| | - Yasuko Tsunetsugu-Yokota
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Medical Technology, School of Human Sciences, Tokyo University of Technology, Tokyo, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan.,Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan.,Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan.,Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
| | - Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
27
|
Izotova N, Rivat C, Baricordi C, Blanco E, Pellin D, Watt E, Gkazi AS, Adams S, Gilmour K, Bayford J, Booth C, Gaspar HB, Thrasher AJ, Biasco L. Long-term lymphoid progenitors independently sustain naïve T and NK cell production in humans. Nat Commun 2021; 12:1622. [PMID: 33712608 PMCID: PMC7954865 DOI: 10.1038/s41467-021-21834-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/06/2021] [Indexed: 12/01/2022] Open
Abstract
Our mathematical model of integration site data in clinical gene therapy supported the existence of long-term lymphoid progenitors capable of surviving independently from hematopoietic stem cells. To date, no experimental setting has been available to validate this prediction. We here report evidence of a population of lymphoid progenitors capable of independently maintaining T and NK cell production for 15 years in humans. The gene therapy patients of this study lack vector-positive myeloid/B cells indicating absence of engineered stem cells but retain gene marking in both T and NK. Decades after treatment, we can still detect and analyse transduced naïve T cells whose production is likely maintained by a population of long-term lymphoid progenitors. By tracking insertional clonal markers overtime, we suggest that these progenitors can support both T and NK cell production. Identification of these long-term lymphoid progenitors could be utilised for the development of next generation gene- and cancer-immunotherapies.
Collapse
Affiliation(s)
- Natalia Izotova
- Great Ormond Street Institute of Child Health Faculty of Population Health Sciences, London, UK
| | - Christine Rivat
- Great Ormond Street Institute of Child Health Faculty of Population Health Sciences, London, UK
- Orchard Therapeutics, University College of London (UCL), London, UK
| | - Cristina Baricordi
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Elena Blanco
- Great Ormond Street Institute of Child Health Faculty of Population Health Sciences, London, UK
| | - Danilo Pellin
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | | | - Athina S Gkazi
- Great Ormond Street Institute of Child Health Faculty of Population Health Sciences, London, UK
| | | | | | | | - Claire Booth
- Great Ormond Street Institute of Child Health Faculty of Population Health Sciences, London, UK
- Great Ormond Street Hospital, London, UK
| | - H Bobby Gaspar
- Great Ormond Street Institute of Child Health Faculty of Population Health Sciences, London, UK
- Orchard Therapeutics, University College of London (UCL), London, UK
| | - Adrian J Thrasher
- Great Ormond Street Institute of Child Health Faculty of Population Health Sciences, London, UK.
- Great Ormond Street Hospital, London, UK.
| | - Luca Biasco
- Great Ormond Street Institute of Child Health Faculty of Population Health Sciences, London, UK.
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Holbrook MC, Goad DW, Grdzelishvili VZ. Expanding the Spectrum of Pancreatic Cancers Responsive to Vesicular Stomatitis Virus-Based Oncolytic Virotherapy: Challenges and Solutions. Cancers (Basel) 2021; 13:1171. [PMID: 33803211 PMCID: PMC7963195 DOI: 10.3390/cancers13051171] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating malignancy with poor prognosis and a dismal survival rate, expected to become the second leading cause of cancer-related deaths in the United States. Oncolytic virus (OV) is an anticancer approach that utilizes replication-competent viruses to preferentially infect and kill tumor cells. Vesicular stomatitis virus (VSV), one such OV, is already in several phase I clinical trials against different malignancies. VSV-based recombinant viruses are effective OVs against a majority of tested PDAC cell lines. However, some PDAC cell lines are resistant to VSV. Upregulated type I IFN signaling and constitutive expression of a subset of interferon-simulated genes (ISGs) play a major role in such resistance, while other mechanisms, such as inefficient viral attachment and resistance to VSV-mediated apoptosis, also play a role in some PDACs. Several alternative approaches have been shown to break the resistance of PDACs to VSV without compromising VSV oncoselectivity, including (i) combinations of VSV with JAK1/2 inhibitors (such as ruxolitinib); (ii) triple combinations of VSV with ruxolitinib and polycations improving both VSV replication and attachment; (iii) combinations of VSV with chemotherapeutic drugs (such as paclitaxel) arresting cells in the G2/M phase; (iv) arming VSV with p53 transgenes; (v) directed evolution approach producing more effective OVs. The latter study demonstrated impressive long-term genomic stability of complex VSV recombinants encoding large transgenes, supporting further clinical development of VSV as safe therapeutics for PDAC.
Collapse
Affiliation(s)
| | | | - Valery Z. Grdzelishvili
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; (M.C.H.); (D.W.G.)
| |
Collapse
|
29
|
Somasundaram R, Connelly T, Choi R, Choi H, Samarkina A, Li L, Gregorio E, Chen Y, Thakur R, Abdel-Mohsen M, Beqiri M, Kiernan M, Perego M, Wang F, Xiao M, Brafford P, Yang X, Xu X, Secreto A, Danet-Desnoyers G, Traum D, Kaestner KH, Huang AC, Hristova D, Wang J, Fukunaga-Kalabis M, Krepler C, Ping-Chen F, Zhou X, Gutierrez A, Rebecca VW, Vonteddu P, Dotiwala F, Bala S, Majumdar S, Dweep H, Wickramasinghe J, Kossenkov AV, Reyes-Arbujas J, Santiago K, Nguyen T, Griss J, Keeney F, Hayden J, Gavin BJ, Weiner D, Montaner LJ, Liu Q, Peiffer L, Becker J, Burton EM, Davies MA, Tetzlaff MT, Muthumani K, Wargo JA, Gabrilovich D, Herlyn M. Tumor-infiltrating mast cells are associated with resistance to anti-PD-1 therapy. Nat Commun 2021; 12:346. [PMID: 33436641 PMCID: PMC7804257 DOI: 10.1038/s41467-020-20600-7] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
Anti-PD-1 therapy is used as a front-line treatment for many cancers, but mechanistic insight into this therapy resistance is still lacking. Here we generate a humanized (Hu)-mouse melanoma model by injecting fetal liver-derived CD34+ cells and implanting autologous thymus in immune-deficient NOD-scid IL2Rγnull (NSG) mice. Reconstituted Hu-mice are challenged with HLA-matched melanomas and treated with anti-PD-1, which results in restricted tumor growth but not complete regression. Tumor RNA-seq, multiplexed imaging and immunohistology staining show high expression of chemokines, as well as recruitment of FOXP3+ Treg and mast cells, in selective tumor regions. Reduced HLA-class I expression and CD8+/Granz B+ T cells homeostasis are observed in tumor regions where FOXP3+ Treg and mast cells co-localize, with such features associated with resistance to anti-PD-1 treatment. Combining anti-PD-1 with sunitinib or imatinib results in the depletion of mast cells and complete regression of tumors. Our results thus implicate mast cell depletion for improving the efficacy of anti-PD-1 therapy. Immune checkpoint therapies (ICT) are promising for treating various cancers, but response rates vary. Here the authors show, in mouse models, that tumor-infiltrating mast cells colocalize with regulatory T cells, coincide with local reduction of MHC-I and CD8 T cells, and is associated with resistance to ICT, which can be reversed by c-kit inhibitor treatment.
Collapse
Affiliation(s)
| | | | - Robin Choi
- The Wistar Institute, Philadelphia, PA, USA
| | | | | | - Ling Li
- The Wistar Institute, Philadelphia, PA, USA
| | | | | | - Rohit Thakur
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | | | | | | | | | - Fang Wang
- The Wistar Institute, Philadelphia, PA, USA
| | - Min Xiao
- The Wistar Institute, Philadelphia, PA, USA
| | | | - Xue Yang
- The Wistar Institute, Philadelphia, PA, USA
| | - Xiaowei Xu
- Department of Pathology and Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anthony Secreto
- Department of Medicine, Stem Cell and Xenograft Core, University of Pennsylvania, Philadelphia, PA, USA
| | - Gwenn Danet-Desnoyers
- Department of Medicine, Stem Cell and Xenograft Core, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Traum
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander C Huang
- Department of Pathology and Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Johannes Griss
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | - Qin Liu
- The Wistar Institute, Philadelphia, PA, USA
| | | | | | - Elizabeth M Burton
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, University of California, San Francisco, CA, USA
| | - Michael T Tetzlaff
- Department of Pathology and Dermatology, University of California, San Francisco, CA, USA
| | - Kar Muthumani
- The Wistar Institute, Philadelphia, PA, USA.,GeneOne Life Science Inc., Fort Washington, PA, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | | | | |
Collapse
|
30
|
Morillon YM, Sabzevari A, Schlom J, Greiner JW. The Development of Next-generation PBMC Humanized Mice for Preclinical Investigation of Cancer Immunotherapeutic Agents. Anticancer Res 2020; 40:5329-5341. [PMID: 32988851 DOI: 10.21873/anticanres.14540] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/14/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
Investigation of the efficacy and mechanisms of human immuno-oncology agents has been hampered due to species-specific differences when utilizing preclinical mouse models. Peripheral blood mononuclear cell (PBMC) humanized mice provide a platform for investigating the modulation of the human immune-mediated antitumor response while circumventing the limitations of syngeneic model systems. Use of humanized mice has been stymied by model-specific limitations, some of which include the development of graft versus host disease, technical difficulty and cost associated with each humanized animal, and insufficient engraftment of some human immune subsets. Recent advances have addressed many of these limitations from which have emerged humanized models that are more clinically relevant. This review characterizes the expanded usage, advantages and limitations of humanized mice and provides insights into the development of the next generation of murine humanized models to further inform clinical applications of cancer immunotherapeutic agents.
Collapse
Affiliation(s)
- Y Maurice Morillon
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Ariana Sabzevari
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, U.S.A.
| | - John W Greiner
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, U.S.A
| |
Collapse
|
31
|
Blümich S, Zdimerova H, Münz C, Kipar A, Pellegrini G. Human CD34 + Hematopoietic Stem Cell-Engrafted NSG Mice: Morphological and Immunophenotypic Features. Vet Pathol 2020; 58:161-180. [PMID: 32901581 DOI: 10.1177/0300985820948822] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunodeficient mice engrafted with human immune cells represent an innovative tool to improve translatability of animal models for the study of human diseases. Immunophenotyping in these mice focuses on engraftment rates and cellular differentiation in blood and secondary lymphoid organs, and is predominantly carried out by FACS (fluorescent activated cell sorting) analysis; information on the morphological aspects of engraftment and the prevalence of histologic lesions is limited. We histologically examined 3- to 6-month-old NSG mice, naïve or engrafted with CD34+ human hemopoietic stem cells (HSC), and employed a quantitative immunohistochemical approach to identify human and murine cell compartments, comparing the results with the FACS data. NSG mice mainly exhibited incidental findings in lungs, kidneys, testes, and adrenal glands. A 6-month-old NSG mouse had a mediastinal lymphoblastic lymphoma. The lymphoid organs of NSG mice lacked typical lymphoid tissue architecture but frequently exhibited small periarteriolar leukocyte clusters in the spleen. Mice engrafted with human HSC frequently showed nephropathy, ovarian atrophy, cataract, and abnormal retinal development, lesions considered secondary to irradiation. In addition, 20% exhibited multisystemic granulomatous inflammatory infiltrates, dominated by human macrophages and T cells, leading to the observed 7% mortality and morbidity. Immunophenotypic data revealed variable repopulation of lymphoid organs with hCD45+ human cells, which did not always parallel the engraftment levels measured via FACS. The study describes the most common pathological features in young NSG mice after human HSC engraftment. As some of these lesions contribute to morbidity, morphological assessment of the engraftment at tissue level might help improve immunophenotypic evaluations of this animal model.
Collapse
Affiliation(s)
- Sandra Blümich
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, 27217University of Zurich, Zurich, Switzerland
| | - Hana Zdimerova
- Viral Immunobiology, Institute of Experimental Immunology, 27217University of Zurich, Zurich, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, 27217University of Zurich, Zurich, Switzerland
| | - Anja Kipar
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, 27217University of Zurich, Zurich, Switzerland
| | - Giovanni Pellegrini
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, 27217University of Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Rijensky NM, Blondheim Shraga NR, Barnea E, Peled N, Rosenbaum E, Popovtzer A, Stemmer SM, Livoff A, Shlapobersky M, Moskovits N, Perry D, Rubin E, Haviv I, Admon A. Identification of Tumor Antigens in the HLA Peptidome of Patient-derived Xenograft Tumors in Mouse. Mol Cell Proteomics 2020; 19:1360-1374. [PMID: 32451349 PMCID: PMC8015002 DOI: 10.1074/mcp.ra119.001876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
Personalized cancer immunotherapy targeting patient-specific cancer/testis antigens (CTA) and neoantigens may benefit from large-scale tumor human leukocyte antigen (HLA) peptidome (immunopeptidome) analysis, which aims to accurately identify antigens presented by tumor cells. Although significant efforts have been invested in analyzing the HLA peptidomes of fresh tumors, it is often impossible to obtain sufficient volumes of tumor tissues for comprehensive HLA peptidome characterization. This work attempted to overcome some of these obstacles by using patient-derived xenograft tumors (PDX) in mice as the tissue sources for HLA peptidome analysis. PDX tumors provide a proxy for the expansion of the patient tumor by re-grafting them through several passages to immune-compromised mice. The HLA peptidomes of human biopsies were compared with those derived from PDX tumors. Larger HLA peptidomes were obtained from the significantly larger PDX tumors as compared with the patient biopsies. The HLA peptidomes of different PDX tumors derived from the same source tumor biopsy were very reproducible, even following subsequent passages to new naïve mice. Many CTA-derived HLA peptides were discovered, as well as several potential neoantigens/variant sequences. Taken together, the use of PDX tumors for HLA peptidome analysis serves as a highly expandable and stable source of reproducible and authentic peptidomes, opening up new opportunities for defining large HLA peptidomes when only small tumor biopsies are available. This approach provides a large source for tumor antigens identification, potentially useful for personalized immunotherapy.
Collapse
Affiliation(s)
| | | | - Eilon Barnea
- Department of Biology, Technion-Israel Institute of Technology Haifa, Israel
| | - Nir Peled
- Institute of Oncology, Davidoff Center, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Eli Rosenbaum
- Institute of Oncology, Davidoff Center, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Aron Popovtzer
- Institute of Oncology, Davidoff Center, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Solomon M Stemmer
- Davidoff Center, Rabin Medical Center, Beilinson Campus, Petach Tikva, and Felsentien medical research center, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alejandro Livoff
- Institute of Pathology, Barzilai University Medical Center, Ashkelon, Israel
| | - Mark Shlapobersky
- Institute of Pathology, Barzilai University Medical Center, Ashkelon, Israel
| | - Neta Moskovits
- Davidoff Center, Rabin Medical Center, Beilinson Campus, Petach Tikva, and Felsentien medical research center, Petach Tikva, Israel
| | - Dafna Perry
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Eitan Rubin
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheva, Israel; The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Itzhak Haviv
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Arie Admon
- Department of Biology, Technion-Israel Institute of Technology Haifa, Israel.
| |
Collapse
|
33
|
Pistollato F, Bernasconi C, McCarthy J, Campia I, Desaintes C, Wittwehr C, Deceuninck P, Whelan M. Alzheimer's Disease, and Breast and Prostate Cancer Research: Translational Failures and the Importance to Monitor Outputs and Impact of Funded Research. Animals (Basel) 2020; 10:E1194. [PMID: 32674379 PMCID: PMC7401638 DOI: 10.3390/ani10071194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022] Open
Abstract
Dementia and cancer are becoming increasingly prevalent in Western countries. In the last two decades, research focused on Alzheimer's disease (AD) and cancer, in particular, breast cancer (BC) and prostate cancer (PC), has been substantially funded both in Europe and worldwide. While scientific research outcomes have contributed to increase our understanding of the disease etiopathology, still the prevalence of these chronic degenerative conditions remains very high across the globe. By definition, no model is perfect. In particular, animal models of AD, BC, and PC have been and still are traditionally used in basic/fundamental, translational, and preclinical research to study human disease mechanisms, identify new therapeutic targets, and develop new drugs. However, animals do not adequately model some essential features of human disease; therefore, they are often unable to pave the way to the development of drugs effective in human patients. The rise of new technological tools and models in life science, and the increasing need for multidisciplinary approaches have encouraged many interdisciplinary research initiatives. With considerable funds being invested in biomedical research, it is becoming pivotal to define and apply indicators to monitor the contribution to innovation and impact of funded research. Here, we discuss some of the issues underlying translational failure in AD, BC, and PC research, and describe how indicators could be applied to retrospectively measure outputs and impact of funded biomedical research.
Collapse
Affiliation(s)
- Francesca Pistollato
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Camilla Bernasconi
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Janine McCarthy
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
- Physicians Committee for Responsible Medicine (PCRM), Washington, DC 20016, USA;
| | - Ivana Campia
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Christian Desaintes
- European Commission, Directorate General for Research and Innovation (RTD), 1000 Brussels, Belgium;
| | - Clemens Wittwehr
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Pierre Deceuninck
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Maurice Whelan
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| |
Collapse
|
34
|
Saito Y, Shultz LD, Ishikawa F. Understanding Normal and Malignant Human Hematopoiesis Using Next-Generation Humanized Mice. Trends Immunol 2020; 41:706-720. [PMID: 32631635 DOI: 10.1016/j.it.2020.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 12/11/2022]
Abstract
Rodent models for human diseases contribute significantly to understanding human physiology and pathophysiology. However, given the accelerating pace of drug development, there is a crucial need for in vivo preclinical models of human biology and pathology. The humanized mouse is one tool to bridge the gap between traditional animal models and the clinic. The development of immunodeficient mouse strains with high-level engraftment of normal and diseased human immune/hematopoietic cells has made in vivo functional characterization possible. As a patient-derived xenograft (PDX) model, humanized mice functionally correlate putative mechanisms with in vivo behavior and help to reveal pathogenic mechanisms. Combined with single-cell genomics, humanized mice can facilitate functional precision medicine such as risk stratification and individually optimized therapeutic approaches.
Collapse
Affiliation(s)
- Yoriko Saito
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, 230-0045, Japan
| | | | - Fumihiko Ishikawa
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, 230-0045, Japan.
| |
Collapse
|
35
|
Hamilton SE, Badovinac VP, Beura LK, Pierson M, Jameson SC, Masopust D, Griffith TS. New Insights into the Immune System Using Dirty Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:3-11. [PMID: 32571979 PMCID: PMC7316151 DOI: 10.4049/jimmunol.2000171] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
The mouse (Mus musculus) is the dominant organism used to investigate the mechanisms behind complex immunological responses because of their genetic similarity to humans and our ability to manipulate those genetics to understand downstream function. Indeed, our knowledge of immune system development, response to infection, and ways to therapeutically manipulate the immune response to combat disease were, in large part, delineated in the mouse. Despite the power of mouse-based immunology research, the translational efficacy of many new therapies from mouse to human is far from ideal. Recent data have highlighted how the naive, neonate-like immune system of specific pathogen-free mice differs dramatically in composition and function to mice living under barrier-free conditions (i.e., "dirty" mice). In this review, we discuss major findings to date and challenges faced when using dirty mice and specific areas of immunology research that may benefit from using animals with robust and varied microbial exposure.
Collapse
Affiliation(s)
- Sara E Hamilton
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA 52242
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Lalit K Beura
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912
| | - Mark Pierson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Stephen C Jameson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
| | - David Masopust
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Thomas S Griffith
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN 55455;
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
- Department of Urology, University of Minnesota, Minneapolis, MN 55455; and
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417
| |
Collapse
|
36
|
Kamdem JP, Duarte AE, Ibrahim M, Lukong KE, Barros LM, Roeder T. Bibliometric analysis of personalized humanized mouse and Drosophila models for effective combinational therapy in cancer patients. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165880. [PMID: 32592936 DOI: 10.1016/j.bbadis.2020.165880] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/25/2020] [Accepted: 06/11/2020] [Indexed: 12/19/2022]
Abstract
Research performed using model organisms such as mice and the fruit fly, Drosophila melanogaster has significantly enhanced our knowledge about cancer biology and the fundamental processes of cancer. This is because the major biological properties and genes associated with cancer including signaling pathways, oncogenes, tumor suppressors, and other regulators of cell growth and proliferation are evolutionary conserved. This review provides bibliometric analysis of research productivity, and performance of authors, institutions, countries, and journals associated with personalized animal cancer models, focussing on the role of Drosophila in cancer research, thus highlighting emerging trends in the field. A total of 1469 and 2672 original articles and reviews for Drosophila cancer model and patient-derived xenograft (PDX) respectively, were retrieved from the Scopus database and the most cited papers were thoroughly analyzed. Our analysis indicates a steadily increasing productivity of the animal models and especially of mouse models in cancer research. In addition to the many different systems that address almost all aspects of tumor research in humanized animal models, a trend towards using tailored screening platforms with Drosophila models in particular will become widespread in the future. Having Drosophila models that recapitulate major genetic aspects of a given tumor will enable the development and validation of novel therapeutic strategies for specific cancers, and provide a platform for screening small molecule inhibitors and other anti-tumor compounds. The combination of Drosophila cancer models and mouse PDX models particularly is highly promising and should be one of the major research strategies the future.
Collapse
Affiliation(s)
- Jean Paul Kamdem
- Department of Biological Sciences, Regional University of Cariri, Campus Pimenta, Crato, Ceara CEP: 63105-000, Brazil.
| | - Antonia Eliene Duarte
- Department of Biological Sciences, Regional University of Cariri, Campus Pimenta, Crato, Ceara CEP: 63105-000, Brazil
| | - Mohammad Ibrahim
- Department of Chemistry, Abdul Wali Khan University Mardan (AWKUM), KPK, Mardan, Pakistan
| | - Kiven Erique Lukong
- Department of Biochemistry, Microbiology and Immunology (BMI) College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada.
| | - Luiz Marivando Barros
- Department of Biological Sciences, Regional University of Cariri, Campus Pimenta, Crato, Ceara CEP: 63105-000, Brazil
| | - Thomas Roeder
- Christian-Albrechts Universität zu Kiel, Zoologisches Institut, Molekulare Physiologie, Olshausenstraße 40, D-24098 Kiel, Germany; German Center for Lung Research, Airway Research Center North, Kiel, Germany.
| |
Collapse
|
37
|
Kummer L, Zaradzki M, Vijayan V, Arif R, Weigand MA, Immenschuh S, Wagner AH, Larmann J. Vascular Signaling in Allogenic Solid Organ Transplantation - The Role of Endothelial Cells. Front Physiol 2020; 11:443. [PMID: 32457653 PMCID: PMC7227440 DOI: 10.3389/fphys.2020.00443] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/09/2020] [Indexed: 12/12/2022] Open
Abstract
Graft rejection remains the major obstacle after vascularized solid organ transplantation. Endothelial cells, which form the interface between the transplanted graft and the host’s immunity, are the first target for host immune cells. During acute cellular rejection endothelial cells are directly attacked by HLA I and II-recognizing NK cells, macrophages, and T cells, and activation of the complement system leads to endothelial cell lysis. The established forms of immunosuppressive therapy provide effective treatment options, but the treatment of chronic rejection of solid organs remains challenging. Chronic rejection is mainly based on production of donor-specific antibodies that induce endothelial cell activation—a condition which phenotypically resembles chronic inflammation. Activated endothelial cells produce chemokines, and expression of adhesion molecules increases. Due to this pro-inflammatory microenvironment, leukocytes are recruited and transmigrate from the bloodstream across the endothelial monolayer into the vessel wall. This mononuclear infiltrate is a hallmark of transplant vasculopathy. Furthermore, expression profiles of different cytokines serve as clinical markers for the patient’s outcome. Besides their effects on immune cells, activated endothelial cells support the migration and proliferation of vascular smooth muscle cells. In turn, muscle cell recruitment leads to neointima formation followed by reduction in organ perfusion and eventually results in tissue injury. Activation of endothelial cells involves antibody ligation to the surface of endothelial cells. Subsequently, intracellular signaling pathways are initiated. These signaling cascades may serve as targets to prevent or treat adverse effects in antibody-activated endothelial cells. Preventive or therapeutic strategies for chronic rejection can be investigated in sophisticated mouse models of transplant vasculopathy, mimicking interactions between immune cells and endothelium.
Collapse
Affiliation(s)
- Laura Kummer
- Department of Anesthesiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Marcin Zaradzki
- Institute of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Vijith Vijayan
- Institute for Transfusion Medicine, Hannover Medical School, Hanover, Germany
| | - Rawa Arif
- Institute of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Markus A Weigand
- Department of Anesthesiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stephan Immenschuh
- Institute for Transfusion Medicine, Hannover Medical School, Hanover, Germany
| | - Andreas H Wagner
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Jan Larmann
- Department of Anesthesiology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
38
|
Sobczuk P, Brodziak A, Khan MI, Chhabra S, Fiedorowicz M, Wełniak-Kamińska M, Synoradzki K, Bartnik E, Cudnoch-Jędrzejewska A, Czarnecka AM. Choosing The Right Animal Model for Renal Cancer Research. Transl Oncol 2020; 13:100745. [PMID: 32092671 PMCID: PMC7036425 DOI: 10.1016/j.tranon.2020.100745] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 12/17/2022] Open
Abstract
The increase in the life expectancy of patients with renal cell carcinoma (RCC) in the last decade is due to changes that have occurred in the area of preclinical studies. Understanding cancer pathophysiology and the emergence of new therapeutic options, including immunotherapy, would not be possible without proper research. Before new approaches to disease treatment are developed and introduced into clinical practice they must be preceded by preclinical tests, in which animal studies play a significant role. This review describes the progress in animal model development in kidney cancer research starting from the oldest syngeneic or chemically-induced models, through genetically modified mice, finally to xenograft, especially patient-derived, avatar and humanized mouse models. As there are a number of subtypes of RCC, our aim is to help to choose the right animal model for a particular kidney cancer subtype. The data on genetic backgrounds, biochemical parameters, histology, different stages of carcinogenesis and metastasis in various animal models of RCC as well as their translational relevance are summarized. Moreover, we shed some light on imaging methods, which can help define tumor microstructure, assist in the analysis of its metabolic changes and track metastasis development.
Collapse
Affiliation(s)
- Paweł Sobczuk
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland; Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland.
| | - Anna Brodziak
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland; Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland.
| | - Mohammed Imran Khan
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada.
| | - Stuti Chhabra
- Department of Biochemistry, CSIR-Central Drug Research Institute, Lucknow, India.
| | - Michał Fiedorowicz
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| | - Marlena Wełniak-Kamińska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| | - Kamil Synoradzki
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| | - Ewa Bartnik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| | - Anna M Czarnecka
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| |
Collapse
|
39
|
Koenig J, Theobald SJ, Stripecke R. Modeling Human Cytomegalovirus in Humanized Mice for Vaccine Testing. Vaccines (Basel) 2020; 8:vaccines8010089. [PMID: 32079250 PMCID: PMC7157227 DOI: 10.3390/vaccines8010089] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/12/2020] [Accepted: 02/12/2020] [Indexed: 12/18/2022] Open
Abstract
Human cytomegalovirus (HCMV or HHV-5) is a globally spread pathogen with strictly human tropism that establishes a life-long persistence. After primary infection, high levels of long-term T and B cell responses are elicited, but the virus is not cleared. HCMV persists mainly in hematopoietic reservoirs, whereby occasional viral reactivation and spread are well controlled in immunocompetent hosts. However, when the immune system cannot control viral infections or reactivations, such as with newborns, patients with immune deficiencies, or immune-compromised patients after transplantations, the lytic outbursts can be severely debilitating or lethal. The development of vaccines for immunization of immune-compromised hosts has been challenging. Several vaccine candidates did not reach the potency expected in clinical trials and were not approved. Before anti-HCMV vaccines can be tested pre-clinically in immune-compromised hosts, reliable in vivo models recapitulating HCMV infection might accelerate their clinical translation. Therefore, immune-deficient mouse strains implanted with human cells and tissues and developing a human immune system (HIS) are being explored to test anti-HCMV vaccines. HIS-mice resemble immune-compromised hosts as they are equipped with antiviral human T and B cells, but the immune reactivity is overall low. Several groups have independently shown that HCMV infections and reactivations can be mirrored in HIS mice. However, these models and the analyses employed varied widely. The path forward is to improve human immune reconstitution and standardize the analyses of adaptive responses so that HIS models can be forthrightly used for testing novel generations of anti-HCMV vaccines in the preclinical pipeline.
Collapse
Affiliation(s)
- Johannes Koenig
- Laboratory of Regenerative Immune Therapies Applied, Excellence Cluster REBIRTH, Hannover Medical School, 30625 Hannover, Germany; (J.K.); (S.J.T.)
- Clinic of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 30625 Hannover, Germany
| | - Sebastian J. Theobald
- Laboratory of Regenerative Immune Therapies Applied, Excellence Cluster REBIRTH, Hannover Medical School, 30625 Hannover, Germany; (J.K.); (S.J.T.)
- Clinic of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 30625 Hannover, Germany
| | - Renata Stripecke
- Laboratory of Regenerative Immune Therapies Applied, Excellence Cluster REBIRTH, Hannover Medical School, 30625 Hannover, Germany; (J.K.); (S.J.T.)
- Clinic of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 30625 Hannover, Germany
- Correspondence: ; Tel.: +49-(511)-532-6999; Fax: +49-(511)-532-6975
| |
Collapse
|
40
|
O’Connell AK, Douam F. Humanized Mice for Live-Attenuated Vaccine Research: From Unmet Potential to New Promises. Vaccines (Basel) 2020; 8:E36. [PMID: 31973073 PMCID: PMC7157703 DOI: 10.3390/vaccines8010036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 01/24/2023] Open
Abstract
Live-attenuated vaccines (LAV) represent one of the most important medical innovations in human history. In the past three centuries, LAV have saved hundreds of millions of lives, and will continue to do so for many decades to come. Interestingly, the most successful LAVs, such as the smallpox vaccine, the measles vaccine, and the yellow fever vaccine, have been isolated and/or developed in a purely empirical manner without any understanding of the immunological mechanisms they trigger. Today, the mechanisms governing potent LAV immunogenicity and long-term induced protective immunity continue to be elusive, and therefore hamper the rational design of innovative vaccine strategies. A serious roadblock to understanding LAV-induced immunity has been the lack of suitable and cost-effective animal models that can accurately mimic human immune responses. In the last two decades, human-immune system mice (HIS mice), i.e., mice engrafted with components of the human immune system, have been instrumental in investigating the life-cycle and immune responses to multiple human-tropic pathogens. However, their use in LAV research has remained limited. Here, we discuss the strong potential of LAVs as tools to enhance our understanding of human immunity and review the past, current and future contributions of HIS mice to this endeavor.
Collapse
Affiliation(s)
| | - Florian Douam
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA 02118, USA;
| |
Collapse
|
41
|
Wunderli SL, Blache U, Snedeker JG. Tendon explant models for physiologically relevant invitro study of tissue biology - a perspective. Connect Tissue Res 2020; 61:262-277. [PMID: 31931633 DOI: 10.1080/03008207.2019.1700962] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Background: Tendon disorders increasingly afflict our aging society but we lack the scientific understanding to clinically address them. Clinically relevant models of tendon disease are urgently needed as established small animal models of tendinopathy fail to capture essential aspects of the disease. Two-dimensional and three-dimensional cell and tissue culture models are similarly limited, lacking many physiological extracellular matrix cues required to maintain tissue homeostasis or guide matrix remodeling. These cues reflect the biochemical and biomechanical status of the tissue, and encode information regarding the mechanical and metabolic competence of the tissue. Tendon explants overcome some of these limitations and have thus emerged as a valuable tool for the discovery and study of mechanisms associated with tendon homeostasis and pathophysiology. Tendon explants retain native cell-cell and cell-matrix connections, while allowing highly reproducible experimental control over extrinsic factors like mechanical loading and nutritional availability. In this sense tendon explant models can deliver insights that are otherwise impossible to obtain from in vivo animal or in vitro cell culture models. Purpose: In this review, we aimed to provide an overview of tissue explant models used in tendon research, with a specific focus on the value of explant culture systems for the controlled study of the tendon core tissue. We discuss their advantages, limitations and potential future utility. We include suggestions and technical recommendations for the successful use of tendon explant cultures and conclude with an outlook on how explant models may be leveraged with state-of-the-art biotechnologies to propel our understanding of tendon physiology and pathology.
Collapse
Affiliation(s)
- Stefania L Wunderli
- University Hospital Balgrist, University of Zurich, Zurich, Switzerland.,Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ulrich Blache
- University Hospital Balgrist, University of Zurich, Zurich, Switzerland.,Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Jess G Snedeker
- University Hospital Balgrist, University of Zurich, Zurich, Switzerland.,Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
42
|
Weichseldorfer M, Heredia A, Reitz M, Bryant JL, Latinovic OS. Use of Humanized Mouse Models for Studying HIV-1 Infection, Pathogenesis and Persistence. JOURNAL OF AIDS AND HIV TREATMENT 2020; 2:23-29. [PMID: 32457941 PMCID: PMC7250391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Despite decades of intensive basic and clinical research efforts, there is still no successful vaccine candidate against human immunodeficiency virus (HIV-1). Standard combined antiretroviral therapy (cART) has been successfully developed and has given remarkable results suppressing HIV-1 infection and transmission. However, cART cannot fully clear the virus from the infected patients. A cure for HIV-1 is highly desirable to stop both the spread of the virus in humans and disease progression in HIV-1 patients. A safe and effective cure strategy for HIV-1 infection will require appropriate animal models that properly mimic HIV-1 infection and advance HIV-1 cure research. Animal models have been a crucial tool in the drug discovery process for investigation of HIV-1 disease mainly in preclinical evaluations of antiretroviral drugs and vaccines. An ideal animal model should recapitulate the main aspects of human-specific HIV-1 infection and pathogenesis with their associated immune responses, while permitting invasive antiretroviral studies. The best humanized mouse models would allow a thorough evaluation of antiretroviral strategies that are aimed towards reducing the establishment and size of the HIV-1 reservoirs. In this review, we evaluate multiple humanized mouse models while presenting their strengths and limitations for HIV-1 research. These humanized mouse models have been tailored in recent decades and heavily employed to address specific quintessential and remaining questions of HIV-1 persistence, pathogenesis and ultimately, eradication.
Collapse
Affiliation(s)
- Matthew Weichseldorfer
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, United States
| | - Alonso Heredia
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, United States,Department of Medicine, School of Medicine, University of Maryland, Baltimore, United States
| | - Marvin Reitz
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, United States,Department of Medicine, School of Medicine, University of Maryland, Baltimore, United States
| | - Joseph L. Bryant
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, United States,Department of Pathology, School of Medicine, University of Maryland, Baltimore, United States
| | - Olga S. Latinovic
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, United States,Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, United States,Correspondence should be addressed to Olga S. Latinovic;
| |
Collapse
|
43
|
Vivier D, Fung K, Rodriguez C, Adumeau P, Ulaner GA, Lewis JS, Sharma SK, Zeglis BM. The Influence of Glycans-Specific Bioconjugation on the FcγRI Binding and In vivo Performance of 89Zr-DFO-Pertuzumab. Am J Cancer Res 2020; 10:1746-1757. [PMID: 32042334 PMCID: PMC6993239 DOI: 10.7150/thno.39089] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022] Open
Abstract
Rationale: The overwhelming majority of radioimmunoconjugates are produced via random conjugation methods predicated on attaching bifunctional chelators to the lysines of antibodies. However, this approach inevitably produces poorly defined and heterogeneous immunoconjugates because antibodies have several lysines distributed throughout their structure. To circumvent this issue, we have previously developed a chemoenzymatic bioconjugation strategy that site-specifically appends cargoes to the biantennary heavy chain glycans attached to CH2 domains of the immunoglobulin's Fc region. In the study at hand, we explore the effects of this approach to site-specific bioconjugation on the Fc receptor binding and in vivo behavior of radioimmunoconjugates. Methods: We synthesized three desferrioxamine (DFO)-labeled immunoconjugates based on the HER2-targeting antibody pertuzumab: one using random bioconjugation methods (DFO-nsspertuzumab) and two using variants of our chemoenzymatic protocol (DFO-sspertuzumab-EndoS and DFO-sspertuzumab-βGal). Subsequently, we characterized these constructs and evaluated their ability to bind HER2, human FcγRI (huFcγRI), and mouse FcγRI (muFcγRI). After radiolabeling the immunoconjugates with zirconium-89, we conducted PET imaging and biodistribution studies in two different mouse models of HER2-expressing breast cancer. Results: MALDI-ToF and SDS-PAGE analysis confirmed the site-specific nature of the bioconjugation, and flow cytometry and surface plasmon resonance (SPR) revealed that all three immunoconjugates bind HER2 as effectively as native pertuzumab. Critically, however, SPR experiments also illuminated that DFO-sspertuzumab-EndoS possesses an attenuated binding affinity for huFcγRI (17.4 ± 0.3 nM) compared to native pertuzumab (4.7 ± 0.2 nM), DFO-nsspertuzumab (4.1 ± 0.1 nM), and DFO-sspertuzumab-βGal (4.7 ± 0.2 nM). ImmunoPET and biodistribution experiments in athymic nude mice bearing HER2-expressing BT474 human breast cancer xenografts yielded no significant differences in the in vivo behavior of the radioimmunoconjugates. Yet experiments in tumor-bearing humanized NSG mice revealed that 89Zr-DFO-sspertuzumab-EndoS produces higher activity concentrations in the tumor (111.8 ± 39.9 %ID/g) and lower activity concentrations in the liver and spleen (4.7 ± 0.8 %ID/g and 13.1 ± 4.0 %ID/g, respectively) than its non-site-specifically labeled cousin, a phenomenon we believe stems from the altered binding of the former to huFcγRI. Conclusion: These data underscore that this approach to site-specific bioconjugation not only produces more homogeneous and well-defined radioimmunoconjugates than traditional methods but may also improve their in vivo performance in mouse models by reducing binding to FcγRI.
Collapse
|
44
|
Curran M, Mairesse M, Matas-Céspedes A, Bareham B, Pellegrini G, Liaunardy A, Powell E, Sargeant R, Cuomo E, Stebbings R, Betts CJ, Saeb-Parsy K. Recent Advancements and Applications of Human Immune System Mice in Preclinical Immuno-Oncology. Toxicol Pathol 2019; 48:302-316. [PMID: 31847725 DOI: 10.1177/0192623319886304] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significant advances in immunotherapies have resulted in the increasing need of predictive preclinical models to improve immunotherapeutic drug development, treatment combination, and to prevent or minimize toxicity in clinical trials. Immunodeficient mice reconstituted with human immune system (HIS), termed humanized mice or HIS mice, permit detailed analysis of human immune biology, development, and function. Although this model constitutes a great translational model, some aspects need to be improved as the incomplete engraftment of immune cells, graft versus host disease and the lack of human cytokines and growth factors. In this review, we discuss current HIS platforms, their pathology, and recent advances in their development to improve the quality of human immune cell reconstitution. We also highlight new technologies that can be used to better understand these models and how improved characterization is needed for their application in immuno-oncology safety, efficacy, and new modalities therapy development.
Collapse
Affiliation(s)
- Michelle Curran
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.,Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Maelle Mairesse
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Alba Matas-Céspedes
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.,Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Bethany Bareham
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Giovanni Pellegrini
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ardi Liaunardy
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Edward Powell
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Rebecca Sargeant
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Emanuela Cuomo
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Richard Stebbings
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Catherine J Betts
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
45
|
Zarrabi M, Afzal E, Asghari MH, Ebrahimi M. Assessment of Short-Term Engraftment Potential of Ex Vivo Expanded Hematopoietic Stem Cells Using Normal Fetal Mouse in Utero Transplantation Model. CELL JOURNAL 2019; 21:259-267. [PMID: 31210431 PMCID: PMC6582413 DOI: 10.22074/cellj.2019.6006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 10/02/2018] [Indexed: 11/04/2022]
Abstract
Objective Ex vivo expansion is a promising strategy to overcome the low number of human umbilical cord blood hematopoietic stem cells (hUCB-HSCs). Although based on the obtained results in unnatural physiological condition of irradiated genetically immune-deficient mouse models, there has always been concern that the expanded cells have less engraftment potential. The purpose of this study was to investigate effect of common ex vivo expansion method on engraftment potential of hUCB-mononuclear cells (MNCs), using normal fetal mouse, as a model with more similarity to human physiological conditions. Materials and Methods In this experimental study, briefly, isolated hUCB-MNCs were cultured in common expansion medium containing stem cell factor, Flt3 ligand and thrombopoietin. The unexpanded and expanded cells were transplanted to the fetal mice on gestational days of 11.5-13.5. After administration of human hematopoiesis growth factors (hHGFs), presence of human CD45+ cells, in the peripheral blood of recipients, was assessed at various time points after transplantation. Results The expanded MNCs showed 32-fold increase in the expression of CD34+38- phenotype and about 3-fold higher clonogenic potential as compared to the uncultured cells. Four weeks after transplantation, 73% (19/26) of expanded-cell recipients and 35% (7/20) of unexpanded-cell recipients were found to be successfully engrafted with human CD45+ cells. The engraftment level of expanded MNCs was significantly (1.8-fold) higher than unexpanded cells. After hHGFs administration, the level was increased to 3.2, 3.8 and 2.6-fold at respectively 8, 12, and 16 weeks of post transplantation. The increased expression of CXCR4 protein in expanded MNCs is a likely explanation for the present findings. Conclusion The presented data showed that expanded MNCs compared to unexpended cells are capable of more rapid and higher short-term engraftment in normal fetal mouse. It could also be suggested that in utero transplantation (IUT) of normal fetal mice could be an appropriate substitute for NOD/SCID mice in xenotransplantation studies.
Collapse
Affiliation(s)
- Morteza Zarrabi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | - Elaheh Afzal
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | - Mohammad Hassan Asghari
- Animal Core Facility, Reproductive Biomedicine Research Center, Royan Institute for Animal Biotechnology, ACECR, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.Electronic Address:
| |
Collapse
|
46
|
Bedognetti D, Ceccarelli M, Galluzzi L, Lu R, Palucka K, Samayoa J, Spranger S, Warren S, Wong KK, Ziv E, Chowell D, Coussens LM, De Carvalho DD, DeNardo DG, Galon J, Kaufman HL, Kirchhoff T, Lotze MT, Luke JJ, Minn AJ, Politi K, Shultz LD, Simon R, Thórsson V, Weidhaas JB, Ascierto ML, Ascierto PA, Barnes JM, Barsan V, Bommareddy PK, Bot A, Church SE, Ciliberto G, De Maria A, Draganov D, Ho WS, McGee HM, Monette A, Murphy JF, Nisticò P, Park W, Patel M, Quigley M, Radvanyi L, Raftopoulos H, Rudqvist NP, Snyder A, Sweis RF, Valpione S, Zappasodi R, Butterfield LH, Disis ML, Fox BA, Cesano A, Marincola FM. Toward a comprehensive view of cancer immune responsiveness: a synopsis from the SITC workshop. J Immunother Cancer 2019; 7:131. [PMID: 31113486 PMCID: PMC6529999 DOI: 10.1186/s40425-019-0602-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/23/2019] [Indexed: 12/13/2022] Open
Abstract
Tumor immunology has changed the landscape of cancer treatment. Yet, not all patients benefit as cancer immune responsiveness (CIR) remains a limitation in a considerable proportion of cases. The multifactorial determinants of CIR include the genetic makeup of the patient, the genomic instability central to cancer development, the evolutionary emergence of cancer phenotypes under the influence of immune editing, and external modifiers such as demographics, environment, treatment potency, co-morbidities and cancer-independent alterations including immune homeostasis and polymorphisms in the major and minor histocompatibility molecules, cytokines, and chemokines. Based on the premise that cancer is fundamentally a disorder of the genes arising within a cell biologic process, whose deviations from normality determine the rules of engagement with the host's response, the Society for Immunotherapy of Cancer (SITC) convened a task force of experts from various disciplines including, immunology, oncology, biophysics, structural biology, molecular and cellular biology, genetics, and bioinformatics to address the complexity of CIR from a holistic view. The task force was launched by a workshop held in San Francisco on May 14-15, 2018 aimed at two preeminent goals: 1) to identify the fundamental questions related to CIR and 2) to create an interactive community of experts that could guide scientific and research priorities by forming a logical progression supported by multiple perspectives to uncover mechanisms of CIR. This workshop was a first step toward a second meeting where the focus would be to address the actionability of some of the questions identified by working groups. In this event, five working groups aimed at defining a path to test hypotheses according to their relevance to human cancer and identifying experimental models closest to human biology, which include: 1) Germline-Genetic, 2) Somatic-Genetic and 3) Genomic-Transcriptional contributions to CIR, 4) Determinant(s) of Immunogenic Cell Death that modulate CIR, and 5) Experimental Models that best represent CIR and its conversion to an immune responsive state. This manuscript summarizes the contributions from each group and should be considered as a first milestone in the path toward a more contemporary understanding of CIR. We appreciate that this effort is far from comprehensive and that other relevant aspects related to CIR such as the microbiome, the individual's recombined T cell and B cell receptors, and the metabolic status of cancer and immune cells were not fully included. These and other important factors will be included in future activities of the taskforce. The taskforce will focus on prioritization and specific actionable approach to answer the identified questions and implementing the collaborations in the follow-up workshop, which will be held in Houston on September 4-5, 2019.
Collapse
Affiliation(s)
| | | | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Université Paris Descartes/Paris V, Paris, France
| | | | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Stefani Spranger
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MT, USA
| | | | - Kwok-Kin Wong
- Perlmutter Cancer Center, New York Langone Health, New York, NY, USA
| | - Elad Ziv
- University of California, San Francisco, San Francisco, CA, USA
| | - Diego Chowell
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Daniel D De Carvalho
- Department of Medical Biophysics, Princess Margaret Cancer Centre University Health Network, University of Toronto, Toronto, Canada
| | - David G DeNardo
- Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Howard L Kaufman
- Massachusetts General Hospital, Boston, MA, USA and Replimune, Inc., Woburn, MA, USA
| | - Tomas Kirchhoff
- Perlmutter Comprehensive Cancer Center, New York University School of Medicine, New York University Langone Health New York, New York, NY, USA
| | - Michael T Lotze
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Andy J Minn
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | - Adrian Bot
- Kite, a Gilead Company, Santa Monica, CA, USA
| | | | | | - Andrea De Maria
- Università degli Studi di Genova and Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | | | - Winson S Ho
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Primary Children's Hospital, University of Utah, Salt Lake City, UT, USA
| | - Heather M McGee
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anne Monette
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | | | - Paola Nisticò
- IRCCS Istituto Nazionale Tumori Regina Elena, Rome, Italy
| | - Wungki Park
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Laszlo Radvanyi
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Nils-Petter Rudqvist
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | | | | | - Sara Valpione
- CRUK Manchester Institute and The Christie NHS Foundation Trust, The University of Manchester, Manchester, UK
| | - Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Bernard A Fox
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Cancer Institute, Portland, OR, USA
| | | | | |
Collapse
|
47
|
Abstract
Perturbed diurnal rhythms are becoming increasingly evident as deleterious events in the pathology of metabolic diseases. Exercise is well characterized as a crucial intervention in the prevention and treatment of individuals with metabolic diseases. Little is known, however, regarding optimizing the timing of exercise bouts in order to maximize their health benefits. Furthermore, exercise is a potent modulator of skeletal muscle metabolism, and it is clear that skeletal muscle has a strong circadian profile. In humans, mitochondrial function peaks in the late afternoon, and the circadian clock might be inherently impaired in myotubes from patients with metabolic disease. Timing exercise bouts to coordinate with an individual's circadian rhythms might be an efficacious strategy to optimize the health benefits of exercise. The role of exercise as a Zeitgeber can also be used as a tool in combating metabolic disease. Shift work is known to induce acute insulin resistance, and appropriately timed exercise might improve health markers in shift workers who are at risk of metabolic disease. In this Review, we discuss the literature regarding diurnal skeletal muscle metabolism and the interaction with exercise bouts at different times of the day to combat metabolic disease.
Collapse
Affiliation(s)
- Brendan M Gabriel
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
- Department of Molecular Medicine and Surgery, Section of Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
48
|
Su H, Cheng Y, Sravanam S, Mathews S, Gorantla S, Poluektova LY, Dash PK, Gendelman HE. Immune Activations and Viral Tissue Compartmentalization During Progressive HIV-1 Infection of Humanized Mice. Front Immunol 2019; 10:340. [PMID: 30873181 PMCID: PMC6403174 DOI: 10.3389/fimmu.2019.00340] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 02/08/2019] [Indexed: 12/12/2022] Open
Abstract
Human immunodeficiency virus type one (HIV-1) tissue compartments are established soon after viral infection. However, the timing in which virus gains a permanent foothold in tissue and the cellular factors that control early viral-immune events are incompletely understood. These are critical events in studies of HIV-1 pathogenesis and in the development of viral reservoirs after antiretroviral therapy. Moreover, factors affecting the permanence of viral-tissue interactions underlie barriers designed to eliminate HIV-1 infection. To this end we investigated the temporal and spatial viral and host factors during HIV-1 seeding of tissue compartments. Two humanized NOD.Cg-Prkdcscid IL2rgtm1Wjl/SzJ mouse models were employed. In the first, immune deficient mice were reconstituted with human CD34+ cord blood hematopoietic stem cells (HSC) (hu-HSC) and in the second mice were transplanted with adult mature human peripheral lymphocytes (hu-PBL). Both, in measure, reflect relationships between immune activation and viral infection as seen in an infected human host. Following humanization both mice models were infected with HIV-1ADA at 104 50% tissue culture infective doses. Viral nucleic acids and protein and immune cell profiles were assayed in brain, lung, spleen, liver, kidney, lymph nodes, bone marrow, and gut from 3 to 42 days. Peripheral CD4+ T cell loss began at 3 days together with detection of HIV-1 RNA in both mouse models after initiation of HIV-1 infection. HIV-1 was observed in all tested tissues at days 3 and 14 in hu- PBL and HSC mice, respectively. Immune impairment was most prominent in hu-PBL mice. T cell maturation and inflammation factors were linked directly to viral tissue seeding in both mouse models. We conclude that early viral tissue compartmentalization provides a roadmap for investigations into HIV-1 elimination.
Collapse
Affiliation(s)
- Hang Su
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Yan Cheng
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sruthi Sravanam
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Saumi Mathews
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Larisa Y. Poluektova
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Prasanta K. Dash
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
49
|
Vivier D, Sharma SK, Adumeau P, Rodriguez C, Fung K, Zeglis BM. The Impact of FcγRI Binding on Immuno-PET. J Nucl Med 2019; 60:1174-1182. [PMID: 30733320 DOI: 10.2967/jnumed.118.223636] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 01/01/2018] [Indexed: 01/07/2023] Open
Abstract
Antibodies are promising vectors for PET imaging. However, the high uptake of radioimmunoconjugates in nontarget tissues such as the liver and spleen hampers their performance as radiotracers. This off-target uptake can lead to suboptimal tumor-to-background activity concentration ratios, decreasing the contrast of images and reducing their diagnostic utility. A possible cause of this uptake is the sequestration of radioimmunoconjugates by immune cells bearing Fc-γ-receptors (FcγR) that bind to the Fc regions of antibodies. Methods: Since the heavy chain glycans influence the affinity of FcγR for the Fc domain, we set out to investigate whether radioimmunoconjugates with truncated glycans would exhibit altered binding to FcγRI and, in turn, improved in vivo performance. Using the HER2-targeting antibody trastuzumab, we synthesized a series of desferrioxamine-bearing immunoconjugates with differing glycosylation states and interrogated their FcγRI binding via surface plasmon resonance, enzyme-linked immunosorbent assay, and flow cytometry. Furthermore, we labeled these immunoconjugates with 89Zr and explored their biodistribution in athymic nude, NSG, and humanized NSG mice bearing human epidermal growth factor receptor 2-expressing human breast cancer xenografts. Results: We observed a strong correlation between the impaired in vitro FcγRI binding of deglycosylated immunoconjugates and significant decreases in the in vivo off-target uptake of the corresponding 89Zr-labeled radioimmunoconjugates (i.e., liver activity concentrations are reduced by ∼3.5-fold in humanized NSG mice). These reductions in off-target uptake were accompanied by concomitant increases in the tumoral activity concentrations of the glycoengineered radioimmunoconjugates, ultimately yielding improved tumor-to-healthy organ contrast and higher quality PET images. Conclusion: Our findings suggest that the deglycosylation of antibodies represents a facile strategy for improving the quality of immuno-PET in animal models as well as in certain patient populations.
Collapse
Affiliation(s)
- Delphine Vivier
- Department of Chemistry, Hunter College of the City University of New York, New York, New York
| | - Sai Kiran Sharma
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Pierre Adumeau
- Department of Chemistry, Hunter College of the City University of New York, New York, New York
| | - Cindy Rodriguez
- Department of Chemistry, Hunter College of the City University of New York, New York, New York
| | - Kimberly Fung
- Department of Chemistry, Hunter College of the City University of New York, New York, New York.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York, New York, New York .,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; and.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York
| |
Collapse
|
50
|
Yao LC, Aryee KE, Cheng M, Kaur P, Keck JG, Brehm MA. Creation of PDX-Bearing Humanized Mice to Study Immuno-oncology. Methods Mol Biol 2019; 1953:241-252. [PMID: 30912026 DOI: 10.1007/978-1-4939-9145-7_15] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A significant obstacle to the study of human cancer biology and the testing of human specific immunotherapeutics is the paucity of translational models that recapitulate both the growth of human tumors and the functionality of human immune systems. Humanized mice engrafted with human hematopoietic stem cells (HSC) and patient-derived xenografts (PDX) enable preclinical investigation of the interactions between the human immune system and human cancer. We use immunodeficient non-obese diabetic (NOD, scid, gamma) NSG™ or NSG™-SGM3 mice as hosts for establishment of human immunity following HSC injection and for engraftment of human tumors. Here we describe a refined protocol for the subcutaneous implant of solid PDX tumors into humanized mice. Protocols to recover infiltrating immune cells from growing tumors and to evaluate the immune cell subsets by flow cytometry are also described.
Collapse
Affiliation(s)
- Li-Chin Yao
- Department of In Vivo Pharmacology Services, The Jackson Laboratory, Sacramento, CA, USA.
| | - Ken-Edwin Aryee
- Department of Molecular Medicine and the Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Mingshan Cheng
- Department of In Vivo Pharmacology Services, The Jackson Laboratory, Sacramento, CA, USA
| | - Pali Kaur
- Department of In Vivo Pharmacology Services, The Jackson Laboratory, Sacramento, CA, USA
| | - James G Keck
- Department of In Vivo Pharmacology Services, The Jackson Laboratory, Sacramento, CA, USA
| | - Michael A Brehm
- Department of Molecular Medicine and the Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|