51
|
Peng Q, Weng K, Li S, Xu R, Wang Y, Wu Y. A Perspective of Epigenetic Regulation in Radiotherapy. Front Cell Dev Biol 2021; 9:624312. [PMID: 33681204 PMCID: PMC7930394 DOI: 10.3389/fcell.2021.624312] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/28/2021] [Indexed: 12/17/2022] Open
Abstract
Radiation therapy (RT) has been employed as a tumoricidal modality for more than 100 years and on 470,000 patients each year in the United States. The ionizing radiation causes genetic changes and results in cell death. However, since the biological mechanism of radiation remains unclear, there is a pressing need to understand this mechanism to improve the killing effect on tumors and reduce the side effects on normal cells. DNA break and epigenetic remodeling can be induced by radiotherapy. Hence the modulation of histone modification enzymes may tune the radiosensitivity of cancer cells. For instance, histone deacetylase (HDAC) inhibitors sensitize irradiated cancer cells by amplifying the DNA damage signaling and inhibiting double-strand DNA break repair to influence the irradiated cells’ survival. However, the combination of epigenetic drugs and radiotherapy has only been evaluated in several ongoing clinical trials for limited cancer types, partly due to a lack of knowledge on the potential mechanisms on how radiation induces epigenetic regulation and chromatin remodeling. Here, we review recent advances of radiotherapy and radiotherapy-induced epigenetic remodeling and introduce related technologies for epigenetic monitoring. Particularly, we exploit the application of fluorescence resonance energy transfer (FRET) biosensors to visualize dynamic epigenetic regulations in single living cells and tissue upon radiotherapy and drug treatment. We aim to bridge FRET biosensor, epigenetics, and radiotherapy, providing a perspective of using FRET to assess epigenetics and provide guidance for radiotherapy to improve cancer treatment. In the end, we discuss the feasibility of a combination of epigenetic drugs and radiotherapy as new approaches for cancer therapeutics.
Collapse
Affiliation(s)
- Qin Peng
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China.,Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States.,Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Kegui Weng
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States.,Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States.,Chongqing Cancer Hospital, Chongqing Cancer Institute, Chongqing University Cancer Hospital, Chongqing, China
| | - Shitian Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States.,Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Richard Xu
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States.,Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Yingxiao Wang
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States.,Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Yongzhong Wu
- Chongqing Cancer Hospital, Chongqing Cancer Institute, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
52
|
Abstract
This note challenges the current idea that a key role of T cells in tumor regression is to directly kill tumor cells. It favors the view that TIL are keys but act indirectly by helping other immune cells to damage the tumor and its stroma.
Collapse
Affiliation(s)
- Nadège Bercovici
- Inserm; U1016; Institut Cochin; Paris, France ; CNRS; UMR8104; Paris, France ; Univ Paris Descartes; Paris, France
| | | |
Collapse
|
53
|
Mahnke YD, Devevre E, Baumgaertner P, Matter M, Rufer N, Romero P, Speiser DE. Human melanoma-specific CD8(+) T-cells from metastases are capable of antigen-specific degranulation and cytolysis directly ex vivo. Oncoimmunology 2021; 1:467-530. [PMID: 22754765 PMCID: PMC3382891 DOI: 10.4161/onci.19856] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The relatively low frequencies of tumor Ag-specific T-cells in PBMC and metastases from cancer patients have long precluded the analysis of their direct ex vivo cytolytic capacity. Using a new composite technique that works well with low cell numbers, we aimed at determining the functional competence of melanoma-specific CD8+ T-cells. A multiparameter flow cytometry based technique was applied to assess the cytolytic function, degranulation and IFNγ production by tumor Ag-specific CD8+ T-cells from PBMC and tumor-infiltrated lymph nodes (TILN) of melanoma patients. We found strong cytotoxicity by T-cells not only when they were isolated from PBMC but also from TILN. Cytotoxicity was observed against peptide-pulsed target cells and melanoma cells presenting the naturally processed endogenous antigen. However, unlike their PBMC-derived counterparts, T-cells from TILN produced only minimal amounts of IFNγ, while exhibiting similar levels of degranulation, revealing a critical functional dichotomy in metastatic lesions. Our finding of partial functional impairment fits well with the current knowledge that T-cells from cancer metastases are so-called exhausted, a state of T-cell hyporesponsiveness also found in chronic viral infections. The identification of responsible mechanisms in the tumor microenvironment is important for improving cancer therapies.
Collapse
Affiliation(s)
- Yolanda D Mahnke
- Ludwig Center for Cancer Research; University of Lausanne; Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
54
|
Grandjean CL, Garcia Z, Lemaître F, Bréart B, Bousso P. Imaging the mechanisms of anti-CD20 therapy in vivo uncovers spatiotemporal bottlenecks in antibody-dependent phagocytosis. SCIENCE ADVANCES 2021; 7:7/8/eabd6167. [PMID: 33608271 PMCID: PMC7895428 DOI: 10.1126/sciadv.abd6167] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 12/31/2020] [Indexed: 05/16/2023]
Abstract
Anti-CD20 antibody (mAb) represents an effective strategy for the treatment of B cell malignancies, possibly involving complement activity, antibody-dependent cellular cytotoxicity and phagocytosis (ADP). While ADP by Kupffer cells deplete circulating tumors, mechanisms targeting non-circulating tumors remain unclear. Using intravital imaging in a model of B cell lymphoma, we establish here the dominance and limitations of ADP in the bone marrow (BM). We found that tumor cells were stably residing in the BM with little evidence for recirculation. To elucidate the mechanism of depletion, we designed a dual fluorescent reporter to visualize phagocytosis and apoptosis. ADP by BM-associated macrophages was the primary mode of tumor elimination but was no longer active after one hour, resulting in partial depletion. Moreover, macrophages were present at low density in tumor-rich regions, targeting only neighboring tumors. Overcoming spatiotemporal bottlenecks in tumor-targeting Ab therapy thus represents a critical path towards the design of optimized therapies.
Collapse
Affiliation(s)
- Capucine L Grandjean
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.
- INSERM U1223, 75015 Paris, France
| | - Zacarias Garcia
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France
- INSERM U1223, 75015 Paris, France
| | - Fabrice Lemaître
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France
- INSERM U1223, 75015 Paris, France
| | - Béatrice Bréart
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France
- INSERM U1223, 75015 Paris, France
| | - Philippe Bousso
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.
- INSERM U1223, 75015 Paris, France
| |
Collapse
|
55
|
Cremasco F, Menietti E, Speziale D, Sam J, Sammicheli S, Richard M, Varol A, Klein C, Umana P, Bacac M, Colombetti S, Perro M. Cross-linking of T cell to B cell lymphoma by the T cell bispecific antibody CD20-TCB induces IFNγ/CXCL10-dependent peripheral T cell recruitment in humanized murine model. PLoS One 2021; 16:e0241091. [PMID: 33406104 PMCID: PMC7787458 DOI: 10.1371/journal.pone.0241091] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022] Open
Abstract
Diffuse large B cell lymphomas (DLBCL) are a highly heterogeneous subtype of Non Hodgkin Lymphoma (NHL), accounting for about 25% of NHL. Despite an increased progression-free survival upon therapy, 40-50% of patients develop relapse/refractory disease, therefore there remains an important medical need. T cell recruiting therapies, such as the CD20xCD3 T cell bi-specific antibody CD20-TCB (RG6026 or glofitamab), represent a novel approach to target all stages of DLBCL, especially those that fail to respond to multiple lines of treatment. We aimed for a better understanding of the molecular features related to the mode of action (MoA) of CD20-TCB in inducing Target/T cell synapse formation and human T cell recruitment to the tumor. To directly evaluate the correlation between synapse, cytokine production and anti-tumor efficacy using CD20-TCB, we developed an innovative preclinical human DLBCL in vivo model that allowed tracking in vivo human T cell dynamics by multiphoton intravital microscopy (MP-IVM). By ex vivo and in vivo approaches, we revealed that CD20-TCB is inducing strong and stable synapses between human T cell and tumor cells, which are dependent on the dose of CD20-TCB and on LFA-1 activity but not on FAS-L. Moreover, despite CD20-TCB being a large molecule (194.342 kDa), we observed that intra-tumor CD20-TCB-mediated human T cell-tumor cell synapses occur within 1 hour upon CD20-TCB administration. These tight interactions, observed for at least 72 hours post TCB administration, result in tumor cell cytotoxicity, resident T cell proliferation and peripheral blood T cell recruitment into tumor. By blocking the IFNγ-CXCL10 axis, the recruitment of peripheral T cells was abrogated, partially affecting the efficacy of CD20-TCB treatment which rely only on resident T cell proliferation. Altogether these data reveal that CD20-TCB's anti-tumor activity relies on a triple effect: i) fast formation of stable T cell-tumor cell synapses which induce tumor cytotoxicity and cytokine production, ii) resident T cell proliferation and iii) recruitment of fresh peripheral T cells to the tumor core to allow a positive enhancement of the anti-tumor effect.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/pharmacology
- Antigens, CD20/immunology
- Antineoplastic Agents, Immunological/pharmacology
- Cell Line, Tumor
- Chemokine CXCL10/immunology
- Humans
- Interferon-gamma/immunology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Mice
- Neoplasm Proteins/immunology
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
| | | | | | - Johannes Sam
- Roche Innovation Center Zürich, Zürich, Switzerland
| | | | | | - Ahmet Varol
- Roche Innovation Center Zürich, Zürich, Switzerland
| | | | - Pablo Umana
- Roche Innovation Center Zürich, Zürich, Switzerland
| | - Marina Bacac
- Roche Innovation Center Zürich, Zürich, Switzerland
| | | | - Mario Perro
- Roche Innovation Center Zürich, Zürich, Switzerland
- * E-mail:
| |
Collapse
|
56
|
Liu L, Dai B, Li R, Liu Z, Zhang Z. Intravital molecular imaging reveals the restrained capacity of CTLs in the killing of tumor cells in the liver. Am J Cancer Res 2021; 11:194-208. [PMID: 33391470 PMCID: PMC7681101 DOI: 10.7150/thno.44979] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 09/16/2020] [Indexed: 01/08/2023] Open
Abstract
Cytotoxic T lymphocytes (CTLs) and their gene-engineered cells display great application prospects in tumor immunotherapy. The timing of CTL-induced molecular events in tumor cells is unclear, and we also unknow whether the killing efficiency of CTLs is restrained in the liver, an immunotolerant organ with a high tumor incidence. Methods: We used intravital imaging to dynamically monitor the fluorescence resonance energy transfer (FRET) signals of caspase-3 and calcium sensor in tumor cells after transferring CTLs into tumor-bearing mice. Results: Our data show that several CTLs attacked on one tumor cell, and on average each CTL killed 1.24 ± 0.11 tumor cells per day in the liver, which was much less efficient than that in the spleen (3.18 ± 0.26 tumor cells/CTL/day). The killing efficiency of CTLs is restrained in the liver and can be reversed by blocking immunosuppressive cytokine. Tumor cells exposed to CTLs appeared to have prolonged calcium influx, which occurred dozens of minutes before caspase-3 activity. Conclusion: The quantitative characterization of these molecular and cellular events provides accurate information to evaluate the efficiency of cellular immunotherapy against tumors and understand the impact of an organ's immune status.
Collapse
|
57
|
Rastogi A, Robert PA, Halle S, Meyer-Hermann M. Evaluation of CD8 T cell killing models with computer simulations of 2-photon imaging experiments. PLoS Comput Biol 2020; 16:e1008428. [PMID: 33370254 PMCID: PMC7793284 DOI: 10.1371/journal.pcbi.1008428] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 01/08/2021] [Accepted: 10/09/2020] [Indexed: 02/01/2023] Open
Abstract
In vivo imaging of cytotoxic T lymphocyte (CTL) killing activity revealed that infected cells have a higher observed probability of dying after multiple contacts with CTLs. We developed a three-dimensional agent-based model to discriminate different hypotheses about how infected cells get killed based on quantitative 2-photon in vivo observations. We compared a constant CTL killing probability with mechanisms of signal integration in CTL or infected cells. The most likely scenario implied increased susceptibility of infected cells with increasing number of CTL contacts where the total number of contacts was a critical factor. However, when allowing in silico T cells to initiate new interactions with apoptotic target cells (zombie contacts), a contact history independent killing mechanism was also in agreement with experimental datasets. The comparison of observed datasets to simulation results, revealed limitations in interpreting 2-photon data, and provided readouts to distinguish CTL killing models.
Collapse
Affiliation(s)
- Ananya Rastogi
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology (BRICS), Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Philippe A. Robert
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology (BRICS), Helmholtz Centre for Infection Research, Braunschweig, Germany
- * E-mail: (PAR); (SH); (MM-H)
| | - Stephan Halle
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- * E-mail: (PAR); (SH); (MM-H)
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology (BRICS), Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
- Centre for Individualised Infection Medicine (CIIM), Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- * E-mail: (PAR); (SH); (MM-H)
| |
Collapse
|
58
|
Upadhyay R, Boiarsky JA, Pantsulaia G, Svensson-Arvelund J, Lin MJ, Wroblewska A, Bhalla S, Scholler N, Bot A, Rossi JM, Sadek N, Parekh S, Lagana A, Baccarini A, Merad M, Brown BD, Brody JD. A Critical Role for Fas-Mediated Off-Target Tumor Killing in T-cell Immunotherapy. Cancer Discov 2020; 11:599-613. [PMID: 33334730 DOI: 10.1158/2159-8290.cd-20-0756] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/11/2020] [Accepted: 11/19/2020] [Indexed: 12/15/2022]
Abstract
T cell-based therapies have induced cancer remissions, though most tumors ultimately progress, reflecting inherent or acquired resistance including antigen escape. Better understanding of how T cells eliminate tumors will help decipher resistance mechanisms. We used a CRISPR/Cas9 screen and identified a necessary role for Fas-FasL in antigen-specific T-cell killing. We also found that Fas-FasL mediated off-target "bystander" killing of antigen-negative tumor cells. This localized bystander cytotoxicity enhanced clearance of antigen-heterogeneous tumors in vivo, a finding that has not been shown previously. Fas-mediated on-target and bystander killing was reproduced in chimeric antigen receptor (CAR-T) and bispecific antibody T-cell models and was augmented by inhibiting regulators of Fas signaling. Tumoral FAS expression alone predicted survival of CAR-T-treated patients in a large clinical trial (NCT02348216). These data suggest strategies to prevent immune escape by targeting both the antigen expression of most tumor cells and the geography of antigen-loss variants. SIGNIFICANCE: This study demonstrates the first report of in vivo Fas-dependent bystander killing of antigen-negative tumors by T cells, a phenomenon that may be contributing to the high response rates of antigen-directed immunotherapies despite tumoral heterogeneity. Small molecules that target the Fas pathway may potentiate this mechanism to prevent cancer relapse.This article is highlighted in the In This Issue feature, p. 521.
Collapse
Affiliation(s)
- Ranjan Upadhyay
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jonathan A Boiarsky
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Gvantsa Pantsulaia
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Judit Svensson-Arvelund
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Matthew J Lin
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aleksandra Wroblewska
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sherry Bhalla
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | - Norah Sadek
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Samir Parekh
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alessandro Lagana
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alessia Baccarini
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Miriam Merad
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Brian D Brown
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joshua D Brody
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York. .,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
59
|
Chitirala P, Chang HF, Martzloff P, Harenberg C, Ravichandran K, Abdulreda MH, Berggren PO, Krause E, Schirra C, Leinders-Zufall T, Benseler F, Brose N, Rettig J. Studying the biology of cytotoxic T lymphocytes in vivo with a fluorescent granzyme B-mTFP knock-in mouse. eLife 2020; 9:e58065. [PMID: 32696761 PMCID: PMC7375811 DOI: 10.7554/elife.58065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/01/2020] [Indexed: 12/23/2022] Open
Abstract
Understanding T cell function in vivo is of key importance for basic and translational immunology alike. To study T cells in vivo, we developed a new knock-in mouse line, which expresses a fusion protein of granzyme B, a key component of cytotoxic granules involved in T cell-mediated target cell-killing, and monomeric teal fluorescent protein from the endogenous Gzmb locus. Homozygous knock-ins, which are viable and fertile, have cytotoxic T lymphocytes with endogeneously fluorescent cytotoxic granules but wild-type-like killing capacity. Expression of the fluorescent fusion protein allows quantitative analyses of cytotoxic granule maturation, transport and fusion in vitro with super-resolution imaging techniques, and two-photon microscopy in living knock-ins enables the visualization of tissue rejection through individual target cell-killing events in vivo. Thus, the new mouse line is an ideal tool to study cytotoxic T lymphocyte biology and to optimize personalized immunotherapy in cancer treatment.
Collapse
Affiliation(s)
- Praneeth Chitirala
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Hsin-Fang Chang
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Paloma Martzloff
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Christiane Harenberg
- Department of Molecular Neurobiology, Max-Planck-Institute of Experimental MedicineGöttingenGermany
| | - Keerthana Ravichandran
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Midhat H Abdulreda
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of MedicineMiamiUnited States
- Department of Surgery, University of Miami Miller School of MedicineMiamiUnited States
- Department of Microbiology and Immunology, University of Miami Miller School of MedicineMiamiUnited States
- Department of Ophthalmology, University of Miami Miller School of MedicineMiamiUnited States
| | - Per-Olof Berggren
- Diabetes Research Institute and Cell Transplant Center, University of Miami Miller School of MedicineMiamiUnited States
- Department of Surgery, University of Miami Miller School of MedicineMiamiUnited States
- Diabetes Research Institute FederationHollywoodUnited States
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Elmar Krause
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Claudia Schirra
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Trese Leinders-Zufall
- Sensory and Neuroendocrine Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| | - Fritz Benseler
- Department of Molecular Neurobiology, Max-Planck-Institute of Experimental MedicineGöttingenGermany
| | - Nils Brose
- Department of Molecular Neurobiology, Max-Planck-Institute of Experimental MedicineGöttingenGermany
| | - Jens Rettig
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland UniversityHomburgGermany
| |
Collapse
|
60
|
Kimm MA, Tzoumas S, Glasl S, Omar M, Symvoulidis P, Olefir I, Rummeny EJ, Meier R, Ntziachristos V. Longitudinal imaging of T cell-based immunotherapy with multi-spectral, multi-scale optoacoustic tomography. Sci Rep 2020; 10:4903. [PMID: 32184401 PMCID: PMC7078227 DOI: 10.1038/s41598-020-61191-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/28/2020] [Indexed: 12/31/2022] Open
Abstract
Most imaging studies of immunotherapy have focused on tracking labeled T cell biodistribution in vivo for understanding trafficking and homing parameters and predicting therapeutic efficacy by the presence of transferred T cells at or in the tumour mass. Conversely, we investigate here a novel concept for longitudinally elucidating anatomical and pathophysiological changes of solid tumours after adoptive T cell transfer in a preclinical set up, using previously unexplored in-tandem macroscopic and mesoscopic optoacoustic (photoacoustic) imaging. We show non-invasive in vivo observations of vessel collapse during tumour rejection across entire tumours and observe for the first time longitudinal tumour rejection in a label-free manner based on optical absorption changes in the tumour mass due to cellular decline. We complement these observations with high resolution episcopic fluorescence imaging of T cell biodistribution using optimized T cell labeling based on two near-infrared dyes targeting the cell membrane and the cytoplasm. We discuss how optoacoustic macroscopy and mesoscopy offer unique contrast and immunotherapy insights, allowing label-free and longitudinal observations of tumour therapy. The results demonstrate optoacoustic imaging as an invaluable tool in understanding and optimizing T cell therapy.
Collapse
Affiliation(s)
- Melanie A Kimm
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stratis Tzoumas
- Chair for Biological Imaging, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Sarah Glasl
- Chair for Biological Imaging, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Murad Omar
- Chair for Biological Imaging, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Panagiotis Symvoulidis
- Chair for Biological Imaging, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ivan Olefir
- Chair for Biological Imaging, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ernst J Rummeny
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Reinhard Meier
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Vasilis Ntziachristos
- Chair for Biological Imaging, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany. .,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
61
|
Hornak I, Rieger H. Stochastic Model of T Cell Repolarization during Target Elimination I. Biophys J 2020; 118:1733-1748. [PMID: 32130873 DOI: 10.1016/j.bpj.2020.01.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/27/2020] [Accepted: 01/30/2020] [Indexed: 12/16/2022] Open
Abstract
Cytotoxic T lymphocytes (T) and natural killer cells are the main cytotoxic killer cells of the human body to eliminate pathogen-infected or tumorigenic cells (i.e., target cells). Once a natural killer or T cell has identified a target cell, they form a tight contact zone, the immunological synapse (IS). One then observes a repolarization of the cell involving the rotation of the microtubule (MT) cytoskeleton and a movement of the MT organizing center (MTOC) to a position that is just underneath the plasma membrane at the center of the IS. Concomitantly, a massive relocation of organelles attached to MTs is observed, including the Golgi apparatus, lytic granules, and mitochondria. Because the mechanism of this relocation is still elusive, we devise a theoretical model for the molecular-motor-driven motion of the MT cytoskeleton confined between plasma membrane and nucleus during T cell polarization. We analyze different scenarios currently discussed in the literature, the cortical sliding and capture-shrinkage mechanisms, and compare quantitative predictions about the spatiotemporal evolution of MTOC position and MT cytoskeleton morphology with experimental observations. The model predicts the experimentally observed biphasic nature of the repositioning due to an interplay between MT cytoskeleton geometry and motor forces and confirms the dominance of the capture-shrinkage over the cortical sliding mechanism when the MTOC and IS are initially diametrically opposed. We also find that the two mechanisms act synergistically, thereby reducing the resources necessary for repositioning. Moreover, it turns out that the localization of dyneins in the peripheral supramolecular activation cluster facilitates their interaction with the MTs. Our model also opens a way to infer details of the dynein distribution from the experimentally observed features of the MT cytoskeleton dynamics. In a subsequent publication, we will address the issue of general initial configurations and situations in which the T cell established two ISs.
Collapse
Affiliation(s)
- Ivan Hornak
- Center for Biophysics (ZBP) and Department of Theoretical Physics, Saarland University, Saarbrücken, Germany
| | - Heiko Rieger
- Center for Biophysics (ZBP) and Department of Theoretical Physics, Saarland University, Saarbrücken, Germany.
| |
Collapse
|
62
|
Li J, Lin W, Chen H, Xu Z, Ye Y, Chen M. Dual-target IL-12-containing nanoparticles enhance T cell functions for cancer immunotherapy. Cell Immunol 2020; 349:104042. [PMID: 32061376 DOI: 10.1016/j.cellimm.2020.104042] [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: 10/16/2019] [Accepted: 01/10/2020] [Indexed: 01/26/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) play a major role in cancer immunotherapy. A potent tumor immunotherapy may not only require activation of anti-tumor effector cells but also rely on the use of cytokines to create a controlled environment for the development of anti-tumor T cells. In this study, we fabricated a dual-target immunonanoparticle, e.g. poly(d,l-lactide-co-glycolide) nanoparticle, by loading Interleukin-12 (IL-12) and modifying with CD8 and Glypican-3 antibodies on the surface. Our results demonstrate that the fabricated targeting immunonanoparticles bind specifically to the two target cells of interest, i.e. CD8+ T cells and HepG-2 cells via the antibody-antigen interactions and form T cell-HepG-2 cell clusters, which enhances the cytotoxicity of T cells. IL-12-containing dual-target immunonanoparticles delivered IL-12 specifically to CD8+ T cells, and favored the expansion, activation and cytotoxic activity of CD8+ T lymphocytes. These results suggest that dual-target IL-12-encapsulated nanoparticles are a promising platform for cancer immunotherapy.
Collapse
Affiliation(s)
- Jieyu Li
- Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China
| | - Wansong Lin
- Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Huijing Chen
- Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China
| | - Zhiping Xu
- Australian Institute for Bioengineering and Nanotechnology (Building 75), The University of Queensland, Cooper Rd., St Lucia, Brisbane, QLD 4072, Australia
| | - Yunbin Ye
- Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China.
| | - Mingshui Chen
- Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China.
| |
Collapse
|
63
|
A flow cytometry-based method to screen for modulators of tumor-specific T cell cytotoxicity. Methods Enzymol 2020; 631:467-482. [PMID: 31948564 DOI: 10.1016/bs.mie.2019.02.040] [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: 03/07/2023]
Abstract
Cancer immunotherapy relies on the ability of immune cells to kill malignant cells. The cytotoxic T lymphocyte (CTL) response is perhaps the functional measure that best reflects cell-mediated immunity against cancer. Straightforward methods that facilitate quantitative evaluation of the potency of compounds that can modulate T cell-mediated, tumor antigen-specific immune responses are central in the screening cascade when searching for new immunotherapeutic agents for cancer. Here we describe a simple, sensitive method, based on flow cytometry analyses, to quantitatively measure cytotoxicity in vitro. We provide examples that validate its feasibility and specificity using CD8+ T lymphocytes specific for a surrogate tumor antigen, and a blocking antibody for the inhibitory PD-1/PD-L1 axis. This method can nonetheless be applied to the screening of virtually any cytotoxicity modulatory compound, including antibodies and small molecules or T cell-based therapies, and can be scaled up for high-throughput workflow and automation.
Collapse
|
64
|
Uppal A, Dehal A, Chang SC, Barrak D, Naeini Y, Jalas JR, Bilchik AJ. The Immune Microenvironment Impacts Survival in Western Patients with Gastric Adenocarcinoma. J Gastrointest Surg 2020; 24:28-38. [PMID: 31625020 DOI: 10.1007/s11605-019-04403-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 09/07/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND Expression of CD3+ T cells, CD8+ cytotoxic T cells, CD45RO+ memory T cells, and FOXP3+ regulatory T cells at the invasive margin (IM) and tumor center (TC) has correlated with survival in gastric adenocarcinoma (GA) patients from East Asia, independent of anatomic staging. The reason for improved survival in East Asians compared with Western patients is a subject of debate. This study examined the immune profiles of a cohort of Western patients with GA, and their association with overall survival (OS). METHODS Immunohistochemistry (IHC) using antibodies to CD3, CD4, CD8, CD45RO, and FOXP3 was performed on a randomly selected resected GA specimens from 88 Western patients. Cutoffs for high or low expression of each marker were determined with maximally selected rank statistics, and multivariable Cox proportional-hazards models constructed to evaluate the relationship between OS and expression of each marker at the IM and TC. RESULTS Immune cell density was independent of anatomic staging. High expression of CD3, CD4, CD8, and CD45RO at the IM along with CD4 and FOXP3 at the TC were associated with improved OS. A combined marker of CD3, CD8, CD45RO, and FOXP3 associated with OS in East Asian GA was also validated. DISCUSSION This is the first report in US patients to demonstrate that high expression of multiple subsets of T lymphocytes in GA is associated with better OS independent of clinical factors and anatomic stage. Further evaluation of immune-modulating mechanisms may explain survival differences between Western and Eastern patients and provide opportunity for novel treatments.
Collapse
Affiliation(s)
- Abhineet Uppal
- Department of Surgical Oncology, John Wayne Cancer Institute, 2200 Santa Monica Blvd, Santa Monica, CA, 90404, USA
| | - Ahmed Dehal
- Department of Surgical Oncology, John Wayne Cancer Institute, 2200 Santa Monica Blvd, Santa Monica, CA, 90404, USA
| | - Shu-Ching Chang
- Medical Data Research Center, Providence St. Joseph Health, Portland, OR, USA
| | - Dany Barrak
- Department of Surgery, Georgetown University Hospital, Washington, DC, USA
| | - Yalda Naeini
- Department of Pathology, Providence St. John's Medical Center, Santa Monica, CA, USA
| | - John R Jalas
- Department of Pathology, Providence St. John's Medical Center, Santa Monica, CA, USA
| | - Anton J Bilchik
- Department of Surgical Oncology, John Wayne Cancer Institute, 2200 Santa Monica Blvd, Santa Monica, CA, 90404, USA.
| |
Collapse
|
65
|
Long-term in vivo microscopy of CAR T cell dynamics during eradication of CNS lymphoma in mice. Proc Natl Acad Sci U S A 2019; 116:24275-24284. [PMID: 31712432 PMCID: PMC6883823 DOI: 10.1073/pnas.1903854116] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Primary central nervous system lymphoma (PCNSL) is a highly malignant brain tumor with limited treatment options. Here, we show that genetically engineered T cells, expressing a chimeric antigen receptor, thoroughly infiltrate these tumors in mice. Combining intravital 2-photon microscopy with chronic cranial windows, we were able to visualize their intratumoral proliferation and intracerebral persistence for up to 159 d, leading to the eradication of large, established PCNSL and to long-term survival. T cells expressing anti-CD19 chimeric antigen receptors (CARs) demonstrate impressive efficacy in the treatment of systemic B cell malignancies, including B cell lymphoma. However, their effect on primary central nervous system lymphoma (PCNSL) is unknown. Additionally, the detailed cellular dynamics of CAR T cells during their antitumor reaction remain unclear, including their intratumoral infiltration depth, mobility, and persistence. Studying these processes in detail requires repeated intravital imaging of precisely defined tumor regions during weeks of tumor growth and regression. Here, we have combined a model of PCNSL with in vivo intracerebral 2-photon microscopy. Thereby, we were able to visualize intracranial PCNSL growth and therapeutic effects of CAR T cells longitudinally in the same animal over several weeks. Intravenous (i.v.) injection resulted in poor tumor infiltration of anti-CD19 CAR T cells and could not sufficiently control tumor growth. After intracerebral injection, however, anti-CD19 CAR T cells invaded deeply into the solid tumor, reduced tumor growth, and induced regression of PCNSL, which was associated with long-term survival. Intracerebral anti-CD19 CAR T cells entered the circulation and infiltrated distant, nondraining lymph nodes more efficiently than mock CAR T cells. After complete regression of tumors, anti-CD19 CAR T cells remained detectable intracranially and intravascularly for up to 159 d. Collectively, these results demonstrate the great potential of anti-CD19 CAR T cells for the treatment of PCNSL.
Collapse
|
66
|
Intravital multiphoton microscopy as a novel tool in the field of immunopharmacology. Pharmacol Ther 2019; 206:107429. [PMID: 31689449 DOI: 10.1016/j.pharmthera.2019.107429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/11/2019] [Indexed: 11/22/2022]
Abstract
Intravital microscopy with multiphoton excitation is a recently developed optical imaging technique for deep tissue imaging without fixation or sectioning, which permits examination of fundamental concepts regarding the dynamic nature of cells under physiological and pathological conditions in living animals. This novel technique also offers exciting opportunities for pharmacological research by providing new platforms for the study of cellular dynamics in response to drugs in vivo. Moreover, fluorescent chemical probes for functional or molecular analysis in single cells in vivo play important roles in pharmacology. For example, we have recently revealed the pharmacodynamic actions of different biological agents for the treatment of rheumatoid arthritis (RA) in vivo by directly visualizing drug-induced cellular behaviors and functions of osteoclasts on bone surfaces. This review focuses on the principles and advantages of intravital imaging for the dissection of pharmacological mechanisms, and discusses how such imaging can contribute to the drug development process, introducing recent trials that evaluated the in vivo pharmacological effects of various agents.
Collapse
|
67
|
Boulch M, Grandjean CL, Cazaux M, Bousso P. Tumor Immunosurveillance and Immunotherapies: A Fresh Look from Intravital Imaging. Trends Immunol 2019; 40:1022-1034. [DOI: 10.1016/j.it.2019.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/12/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022]
|
68
|
Khazen R, Müller S, Lafouresse F, Valitutti S, Cussat-Blanc S. Sequential adjustment of cytotoxic T lymphocyte densities improves efficacy in controlling tumor growth. Sci Rep 2019; 9:12308. [PMID: 31444380 PMCID: PMC6707257 DOI: 10.1038/s41598-019-48711-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/09/2019] [Indexed: 12/30/2022] Open
Abstract
Understanding the human cytotoxic T lymphocyte (CTL) biology is crucial to develop novel strategies aiming at maximizing their lytic capacity against cancer cells. Here we introduce an agent-based model, calibrated on population-scale experimental data that allows quantifying human CTL per capita killing. Our model highlights higher individual CTL killing capacity at lower CTL densities and fits experimental data of human melanoma cell killing. The model allows extending the analysis over prolonged time frames, difficult to investigate experimentally, and reveals that initial high CTL densities hamper efficacy to control melanoma growth. Computational analysis forecasts that sequential addition of fresh CTL cohorts improves tumor growth control. In vivo experimental data, obtained in a mouse melanoma model, confirm this prediction. Taken together, our results unveil the impact that sequential adjustment of cellular densities has on enhancing CTL efficacy over long-term confrontation with tumor cells. In perspective, they can be instrumental to refine CTL-based therapeutic strategies aiming at controlling tumor growth.
Collapse
Affiliation(s)
- Roxana Khazen
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM/ Université Toulouse III Paul Sabatier, «Equipe labellisée Ligue Nationale contre le cancer 2018», INSERM, Toulouse, France.,INSERM U1223, Dynamics of Immune Responses Unit, Institut Pasteur, 75015, Paris, France
| | - Sabina Müller
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM/ Université Toulouse III Paul Sabatier, «Equipe labellisée Ligue Nationale contre le cancer 2018», INSERM, Toulouse, France
| | - Fanny Lafouresse
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM/ Université Toulouse III Paul Sabatier, «Equipe labellisée Ligue Nationale contre le cancer 2018», INSERM, Toulouse, France
| | - Salvatore Valitutti
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM/ Université Toulouse III Paul Sabatier, «Equipe labellisée Ligue Nationale contre le cancer 2018», INSERM, Toulouse, France. .,Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, 31059, Toulouse, France.
| | - Sylvain Cussat-Blanc
- Institute of Advanced Technologies in Living Sciences, CNRS - USR3505, Toulouse, France.,University of Toulouse, Institute of Research in Informatics of Toulouse, CNRS - UMR5505, Toulouse, France
| |
Collapse
|
69
|
Milo I, Bedora-Faure M, Garcia Z, Thibaut R, Périé L, Shakhar G, Deriano L, Bousso P. The immune system profoundly restricts intratumor genetic heterogeneity. Sci Immunol 2019; 3:3/29/eaat1435. [PMID: 30470696 DOI: 10.1126/sciimmunol.aat1435] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 08/03/2018] [Accepted: 10/18/2018] [Indexed: 12/14/2022]
Abstract
Tumors develop under the selective pressure of the immune system. However, it remains critical to establish how the immune system affects the clonal heterogeneity of tumors that often display cell-to-cell variation in genetic alterations and antigenic expression. To address these questions, we introduced a multicolor barcoding strategy to study the growth of a MYC-driven B cell lymphoma harboring a large degree of intratumor genetic diversity. Using intravital imaging, we visualized that lymphoma subclones grow as patches of sessile cells in the bone marrow, creating a spatially compartmentalized architecture for tumor diversity. Using multicolor barcoding and whole-exome sequencing, we demonstrated that immune responses strongly restrict intratumor genomic diversity and favor clonal dominance, a process mediated by the selective elimination of more immunogenic cells and amplified by epitope spreading. Anti-PD-1 treatment also narrowed intratumor diversity. Our results provide direct evidence that immune pressure shapes the level of intratumor genetic heterogeneity and have important implications for the design of therapeutic strategies.
Collapse
Affiliation(s)
- Idan Milo
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.,INSERM U1223, 75015 Paris, France
| | - Marie Bedora-Faure
- Genome Integrity, Immunity and Cancer Unit, Department of Immunology, Department of Genomes and Genetics, Institut Pasteur, 75015 Paris, France
| | - Zacarias Garcia
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.,INSERM U1223, 75015 Paris, France
| | - Ronan Thibaut
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.,INSERM U1223, 75015 Paris, France.,University Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, rue du Dr Roux, 75015 Paris, France
| | - Leïla Périé
- Institut Curie, PSL Research University, CNRS UMR168, 11 rue Pierre et Marie Curie, 75005 Paris, France.,Sorbonne Universités, UPMC University Paris 06, 4 place Jussieu, 75005 Paris, France
| | - Guy Shakhar
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ludovic Deriano
- Genome Integrity, Immunity and Cancer Unit, Department of Immunology, Department of Genomes and Genetics, Institut Pasteur, 75015 Paris, France
| | - Philippe Bousso
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France. .,INSERM U1223, 75015 Paris, France
| |
Collapse
|
70
|
Manaster Y, Shipony Z, Hutzler A, Kolesnikov M, Avivi C, Shalmon B, Barshack I, Besser MJ, Feferman T, Shakhar G. Reduced CTL motility and activity in avascular tumor areas. Cancer Immunol Immunother 2019; 68:1287-1301. [PMID: 31253998 PMCID: PMC11028152 DOI: 10.1007/s00262-019-02361-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 06/16/2019] [Indexed: 01/04/2023]
Abstract
Patchy infiltration of tumors by cytotoxic T cells (CTLs) predicts poorer prognosis for cancer patients. The factors limiting intratumoral CTL dissemination, though, are poorly understood. To study CTL dissemination in tumors, we histologically examined human melanoma samples and used mice to image B16-OVA tumors infiltrated by OT-I CTLs using intravital two-photon microscopy. In patients, most CTLs concentrated around peripheral blood vessels, especially in poorly infiltrated tumors. In mice, OT-I CTLs had to cluster around tumor cells to efficiently kill them in a contact-and perforin-dependent manner and cytotoxicity was strictly antigen-specific. OT-I CTLs as well as non-specific CTLs concentrated around peripheral vessels, and cleared the tumor cells around them. This was also the case when CTLs were injected directly into the tumors. CTLs crawled rapidly only in areas within 50 µm of flowing blood vessels and transient occlusion of vessels immediately, though reversibly, stopped their migration. In vitro, oxygen depletion and blockade of oxidative phosphorylation also reduced CTL motility. Taken together, these results suggest that hypoxia limits CTL migration away from blood vessels, providing immune-privileged niches for tumor cells to survive. Normalizing intratumoral vasculature may thus synergize with tumor immunotherapy.
Collapse
Affiliation(s)
- Yoav Manaster
- Department of Immunology, Weizmann Institute of Science, Wolfson Bldg., 234 Herzl St., 76100, Rehovot, Israel
| | - Zohar Shipony
- Department of Immunology, Weizmann Institute of Science, Wolfson Bldg., 234 Herzl St., 76100, Rehovot, Israel
| | - Anat Hutzler
- Department of Immunology, Weizmann Institute of Science, Wolfson Bldg., 234 Herzl St., 76100, Rehovot, Israel
| | - Masha Kolesnikov
- Department of Immunology, Weizmann Institute of Science, Wolfson Bldg., 234 Herzl St., 76100, Rehovot, Israel
| | - Camila Avivi
- Department of Pathology, Sheba Medical Center, Derech Sheba 2, 52621, Ramat Gan, Israel
| | - Bruria Shalmon
- Department of Pathology, Sheba Medical Center, Derech Sheba 2, 52621, Ramat Gan, Israel
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Derech Sheba 2, 52621, Ramat Gan, Israel
| | - Michal J Besser
- Ella Institute, Sheba Medical Center, Derech Sheba 2, 52621, Ramat Gan, Israel
- Department of Human Microbiology and Immunology, Sackler Medical School, Tel-Aviv University, 35 Klachkin st, 6997801, Tel Aviv, Israel
| | - Tali Feferman
- Department of Immunology, Weizmann Institute of Science, Wolfson Bldg., 234 Herzl St., 76100, Rehovot, Israel.
| | - Guy Shakhar
- Department of Immunology, Weizmann Institute of Science, Wolfson Bldg., 234 Herzl St., 76100, Rehovot, Israel.
| |
Collapse
|
71
|
Minoshima M, Kikuta J, Omori Y, Seno S, Suehara R, Maeda H, Matsuda H, Ishii M, Kikuchi K. In Vivo Multicolor Imaging with Fluorescent Probes Revealed the Dynamics and Function of Osteoclast Proton Pumps. ACS CENTRAL SCIENCE 2019; 5:1059-1066. [PMID: 31263765 PMCID: PMC6598158 DOI: 10.1021/acscentsci.9b00220] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Indexed: 05/05/2023]
Abstract
In vivo two-photon fluorescence imaging is a powerful modality to monitor cell dynamics in biomedical studies. To detect protein functions in living animals in real-time, fluorescent probes must show a quick response to the target function in specific tissues. Here, we developed a rhodamine-based small-molecule fluorescent probe called Red-pHocas (red pH-activatable fluorescent probe for osteoclast activity sensing) to reversibly detect the acidic environments for the spatiotemporal analysis of the function of osteoclast proton pumps. The introduction of electron-withdrawing N-alkyl substituents in the rhodamine spirolactam fluorophore remarkably increased the kinetics of the fluorescence response to acidic pHs, which allowed the rapid and reversible monitoring of acidic compartments and the analysis of the dynamics of osteoclast proton pumps during osteoclastic bone resorption. In vivo multicolor two-photon imaging using Red-pHocas in fluorescent reporter mice revealed that bone acidification occurred synchronously with the accumulation of proton pumps onto the bone surfaces. To our knowledge, this is the first study to demonstrate the direct involvement of osteoclast proton pumps in bone acidification under intravital conditions by means of an imaging probe.
Collapse
Affiliation(s)
- Masafumi Minoshima
- Department
of Material and Life Science, Graduate School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan
| | - Junichi Kikuta
- Department
of Immunology and Cell Biology, Graduate School of Medicine and Frontier
Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
- WPI—Immunology
Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yuta Omori
- Department
of Material and Life Science, Graduate School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan
| | - Shigeto Seno
- Department
of Bioinformatic Engineering, Graduate School of Information Science
and Technology, Osaka University, Suita, Osaka 565-0871, Japan
| | - Riko Suehara
- Department
of Immunology and Cell Biology, Graduate School of Medicine and Frontier
Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroki Maeda
- Department
of Material and Life Science, Graduate School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hideo Matsuda
- Department
of Bioinformatic Engineering, Graduate School of Information Science
and Technology, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masaru Ishii
- Department
of Immunology and Cell Biology, Graduate School of Medicine and Frontier
Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
- WPI—Immunology
Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kazuya Kikuchi
- Department
of Material and Life Science, Graduate School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan
- WPI—Immunology
Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
- E-mail:
| |
Collapse
|
72
|
Beck RJ, Slagter M, Beltman JB. Contact-Dependent Killing by Cytotoxic T Lymphocytes Is Insufficient for EL4 Tumor Regression In Vivo. Cancer Res 2019; 79:3406-3416. [PMID: 31040155 DOI: 10.1158/0008-5472.can-18-3147] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 02/08/2019] [Accepted: 04/25/2019] [Indexed: 11/16/2022]
Abstract
Immunotherapies are an emerging strategy for treatment of solid tumors. Improved understanding of the mechanisms employed by cytotoxic T lymphocytes (CTL) to control tumors will aid in the development of immunotherapies. CTLs can directly kill tumor cells in a contact-dependent manner or may exert indirect effects on tumor cells via secretion of cytokines. Here, we aim to quantify the importance of these mechanisms in murine thymoma EL4/EG7 cells. We developed an agent-based model (ABM) and an ordinary differential equation model of tumor regression after adoptive transfer of a population of CTLs. Models were parameterized based on in vivo measurements of CTL infiltration and killing rates applied to EL4/EG7 tumors and OTI T cells. We quantified whether infiltrating CTLs are capable of controlling tumors through only direct, contact-dependent killing. Both models agreed that the low measured killing rate of CTLs in vivo was insufficient to cause tumor regression. In our ABM, we also simulated CTL production of the cytokine IFNγ in order to explore how an antiproliferative effect of IFNγ might aid CTLs in tumor control. In this model, IFNγ substantially reduced tumor growth compared with direct killing alone. Collectively, these data demonstrate that contact-dependent killing is insufficient for EL4 regression in vivo and highlight the potential importance of cytokine-induced antiproliferative effects in T-cell-mediated tumor control. SIGNIFICANCE: Computational modeling highlights the importance of cytokine-induced antiproliferative effects in T-cell-mediated control of tumor progression.
Collapse
Affiliation(s)
- Richard J Beck
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Maarten Slagter
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Joost B Beltman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
73
|
Hyun YM, Choe YH, Park SA, Kim M. LFA-1 (CD11a/CD18) and Mac-1 (CD11b/CD18) distinctly regulate neutrophil extravasation through hotspots I and II. Exp Mol Med 2019; 51:1-13. [PMID: 30967528 PMCID: PMC6456621 DOI: 10.1038/s12276-019-0227-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/21/2018] [Accepted: 12/11/2018] [Indexed: 01/21/2023] Open
Abstract
Precise spatiotemporal regulation of leukocyte extravasation is key for generating an efficient immune response to injury or infection. The integrins LFA-1(CD11a/CD18) and Mac-1(CD11b/CD18) play overlapping roles in neutrophil migration because they bind the same as well as different ligands in response to extracellular signaling. Using two-photon intravital imaging and transmission electron microscopy, we observed the existence of preferred sites for neutrophil entrance into the endothelial cell monolayer and exit from the basement membrane and pericyte sheath during neutrophil extravasation, namely, hotspots I and II, by elucidating distinctive roles of LFA-1 and Mac-1. To penetrate the vascular endothelium, neutrophils must first penetrate the endothelial cell layer through hotspot I (i.e., the point of entry into the endothelium). Neutrophils frequently remain in the space between the endothelial cell layer and the basement membrane for a prolonged period (>20 min). Subsequently, neutrophils penetrate the basement membrane and pericyte sheath at hotspot II, which is the final stage of exiting the vascular endothelium. To further investigate the roles of LFA-1 and Mac-1, we newly generated LFA-1 FRET (CD11a-YFP/CD18-CFP) mice and Mac-1 FRET (CD11b-YFP/CD18-CFP) mice. Using both FRET mice, we were able to determine that LFA-1 and Mac-1 distinctly regulate the neutrophil extravasation cascade. Our data suggest that the vascular endothelium functions as a double-layered barrier in the steps of neutrophil extravasation. We propose that the harmonized regulation of neutrophil penetration through the endothelium via hotspots I and II may be critical for vascular homeostasis during inflammation.
Collapse
Affiliation(s)
- Young-Min Hyun
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Young Ho Choe
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang A Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- School of Medicine, CHA University, Seongnam, South Korea
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| |
Collapse
|
74
|
Cazaux M, Grandjean CL, Lemaître F, Garcia Z, Beck RJ, Milo I, Postat J, Beltman JB, Cheadle EJ, Bousso P. Single-cell imaging of CAR T cell activity in vivo reveals extensive functional and anatomical heterogeneity. J Exp Med 2019; 216:1038-1049. [PMID: 30936262 PMCID: PMC6504219 DOI: 10.1084/jem.20182375] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/07/2019] [Accepted: 03/04/2019] [Indexed: 12/21/2022] Open
Abstract
Cazaux et al. use intravital imaging to dissect anti-CD19 CAR T cell activity. This study uncovers both anatomical and functional diversity in the outcome of anti-CD19 CAR T cell interactions with tumor cells impacting engraftment, killing dynamics, and tumor immunoediting. CAR T cells represent a potentially curative strategy for B cell malignancies. However, the outcome and dynamics of CAR T cell interactions in distinct anatomical sites are poorly understood. Using intravital imaging, we tracked interactions established by anti-CD19 CAR T cells in B cell lymphoma–bearing mice. Circulating targets trapped CAR T cells in the lungs, reducing their access to lymphoid organs. In the bone marrow, tumor apoptosis was largely due to CAR T cells that engaged, killed, and detached from their targets within 25 min. Notably, not all CAR T cell contacts elicited calcium signaling or killing while interacting with tumors, uncovering extensive functional heterogeneity. Mathematical modeling revealed that direct killing was sufficient for tumor regression. Finally, antigen-loss variants emerged in the bone marrow, but not in lymph nodes, where CAR T cell cytotoxic activity was reduced. Our results identify a previously unappreciated level of diversity in the outcomes of CAR T cell interactions in vivo, with important clinical implications.
Collapse
Affiliation(s)
- Marine Cazaux
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, INSERM U1223, Paris, France.,University Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Capucine L Grandjean
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, INSERM U1223, Paris, France
| | - Fabrice Lemaître
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, INSERM U1223, Paris, France
| | - Zacarias Garcia
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, INSERM U1223, Paris, France
| | - Richard J Beck
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Idan Milo
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, INSERM U1223, Paris, France
| | - Jérémy Postat
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, INSERM U1223, Paris, France.,University Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Joost B Beltman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Eleanor J Cheadle
- Targeted Therapy Group, Manchester Cancer Research Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Philippe Bousso
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, INSERM U1223, Paris, France
| |
Collapse
|
75
|
Thompson EA, Mitchell JS, Beura LK, Torres DJ, Mrass P, Pierson MJ, Cannon JL, Masopust D, Fife BT, Vezys V. Interstitial Migration of CD8αβ T Cells in the Small Intestine Is Dynamic and Is Dictated by Environmental Cues. Cell Rep 2019; 26:2859-2867.e4. [PMID: 30865878 PMCID: PMC6754515 DOI: 10.1016/j.celrep.2019.02.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 08/05/2018] [Accepted: 02/08/2019] [Indexed: 02/08/2023] Open
Abstract
The migratory capacity of adaptive CD8αβ T cells dictates their ability to locate target cells and exert cytotoxicity, which is the basis of immune surveillance for the containment of microbes and disease. The small intestine (SI) is the largest mucosal surface and is a primary site of pathogen entrance. Using two-photon laser scanning microscopy, we found that motility of antigen (Ag)-specific CD8αβ T cells in the SI is dynamic and varies with the environmental milieu. Pathogen-specific CD8αβ T cell movement differed throughout infection, becoming locally confined at memory. Motility was not dependent on CD103 but was influenced by micro-anatomical locations within the SI and by inflammation. CD8 T cells responding to self-protein were initially affected by the presence of self-Ag, but this was altered after complete tolerance induction. These studies identify multiple factors that affect CD8αβ T cell movement in the intestinal mucosa and show the adaptability of CD8αβ T cell motility.
Collapse
Affiliation(s)
- Emily A Thompson
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jason S Mitchell
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lalit K Beura
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - David J Torres
- Department of Mathematics and Physical Science, Northern New Mexico College, Espanola, NM 87532, USA
| | - Paulus Mrass
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Mark J Pierson
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Judy L Cannon
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM 87131, USA
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Brian T Fife
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Vaiva Vezys
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
76
|
Qi S, Shi H, Liu L, Zhou L, Zhang Z. Dynamic visualization of the whole process of cytotoxic T lymphocytes killing B16 tumor cells in vitro. JOURNAL OF BIOMEDICAL OPTICS 2019; 24:1-7. [PMID: 30825298 PMCID: PMC6992963 DOI: 10.1117/1.jbo.24.5.051413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/14/2019] [Indexed: 06/09/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) play a key role in adoptive cell therapy (ACT) by destroying tumor cells. Although some mechanisms of CTLs killing tumor cells have already been revealed, the precise dynamic information of CTLs' interaction with tumor cells is still not known. Here, we used confocal microscopy to visualize the whole process of how CTLs kill tumor cells in vitro. According to imaging data, CTLs destroyed the target tumor cells rapidly and efficiently. Several CTLs surrounded one or more tumor cells, and the average time for CTLs destroying one or more tumor cells in vitro is dozens of minutes only. Our study displayed the temporal events of CTLs' interaction with tumor cells at the beginning up to the point of killing them. Furthermore, the imaging data presented strong cytotoxicity of CTLs toward the specific tumor cells. These results could help us to well understand the mechanism of CTLs' elimination of tumor cells and improve the efficacy of ACT in cancer immunotherapy.
Collapse
Affiliation(s)
- Shuhong Qi
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hua Shi
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lili Zhou
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhihong Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
77
|
Naumenko V, Van S, Dastidar H, Kim DS, Kim SJ, Zeng Z, Deniset J, Lau A, Zhang C, Macia N, Heyne B, Jenne CN, Mahoney DJ. Visualizing Oncolytic Virus-Host Interactions in Live Mice Using Intravital Microscopy. MOLECULAR THERAPY-ONCOLYTICS 2018; 10:14-27. [PMID: 30073187 PMCID: PMC6070694 DOI: 10.1016/j.omto.2018.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/05/2018] [Indexed: 12/13/2022]
Abstract
Oncolytic virus (OV) therapy is an emerging cancer treatment that uses replicating viruses to infect and kill tumor cells and incite anticancer immunity. While the approach shows promise, it currently fails most patients, indicating strategies to improve OV activity are needed. Developing these will require greater understanding of OV biology, particularly in the context of OV delivery and clearance, the infection process within a complex tumor microenvironment, and the modulation of anticancer immunity. To help achieve this, we have established a technique for high-resolution 4D imaging of OV-host interactions within intact tissues of live mice using intravital microscopy (IVM). We show that oncolytic vesicular stomatitis virus (VSV) directly labeled with Alexa Fluor dyes is easily visualized by single- or multiphoton microscopy while retaining bioactivity in vivo. The addition of fluorophore-tagged antibodies and genetically encoded reporter proteins to image target cells and the virus infection enables real-time imaging of dynamic interactions between VSV and host cells in blood, tumor, and visceral organs of live mice. The method has sufficient in vivo resolution to observe leukocytes in blood binding to and transporting VSV particles, foci of VSV infection spreading through a tumor, and antigen-presenting cells in the spleen interacting with and being infected by VSV. Visualizing OV-host interactions by IVM represents a powerful new tool for studying OV therapy.
Collapse
Affiliation(s)
- Victor Naumenko
- Alberta Children's Hospital Research Institute, Calgary, AB T2N 4N1, Canada.,Arnie Charbonneau Cancer Institute, Calgary, AB T2N 4N1, Canada.,Snyder Institute for Chronic Disease, Calgary, AB T2N 4N1, Canada.,National University of Science and Technology "MISIS," Leninskiy prospect 4, 119991 Moscow, Russia
| | - Shinia Van
- Alberta Children's Hospital Research Institute, Calgary, AB T2N 4N1, Canada.,Arnie Charbonneau Cancer Institute, Calgary, AB T2N 4N1, Canada.,Snyder Institute for Chronic Disease, Calgary, AB T2N 4N1, Canada
| | - Himika Dastidar
- Alberta Children's Hospital Research Institute, Calgary, AB T2N 4N1, Canada.,Arnie Charbonneau Cancer Institute, Calgary, AB T2N 4N1, Canada.,Department of Microbiology, Immunology and Infectious Disease, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Dae-Sun Kim
- Alberta Children's Hospital Research Institute, Calgary, AB T2N 4N1, Canada.,Arnie Charbonneau Cancer Institute, Calgary, AB T2N 4N1, Canada
| | - Seok-Joo Kim
- Snyder Institute for Chronic Disease, Calgary, AB T2N 4N1, Canada
| | - Zhutian Zeng
- Snyder Institute for Chronic Disease, Calgary, AB T2N 4N1, Canada
| | - Justin Deniset
- Snyder Institute for Chronic Disease, Calgary, AB T2N 4N1, Canada
| | - Arthur Lau
- Department of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Chunfen Zhang
- Alberta Children's Hospital Research Institute, Calgary, AB T2N 4N1, Canada.,Arnie Charbonneau Cancer Institute, Calgary, AB T2N 4N1, Canada
| | - Nicolas Macia
- Department of Chemistry, Faculty of Science, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Belinda Heyne
- Department of Chemistry, Faculty of Science, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Craig N Jenne
- Snyder Institute for Chronic Disease, Calgary, AB T2N 4N1, Canada.,Department of Microbiology, Immunology and Infectious Disease, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Douglas J Mahoney
- Alberta Children's Hospital Research Institute, Calgary, AB T2N 4N1, Canada.,Arnie Charbonneau Cancer Institute, Calgary, AB T2N 4N1, Canada.,Department of Microbiology, Immunology and Infectious Disease, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.,Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
78
|
Visualizing Viral Infection In Vivo by Multi-Photon Intravital Microscopy. Viruses 2018; 10:v10060337. [PMID: 29925766 PMCID: PMC6024644 DOI: 10.3390/v10060337] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/12/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022] Open
Abstract
Viral pathogens have adapted to the host organism to exploit the cellular machinery for virus replication and to modulate the host cells for efficient systemic dissemination and immune evasion. Much of our knowledge of the effects that virus infections have on cells originates from in vitro imaging studies using experimental culture systems consisting of cell lines and primary cells. Recently, intravital microscopy using multi-photon excitation of fluorophores has been applied to observe virus dissemination and pathogenesis in real-time under physiological conditions in living organisms. Critical steps during viral infection and pathogenesis could be studied by direct visualization of fluorescent virus particles, virus-infected cells, and the immune response to viral infection. In this review, I summarize the latest research on in vivo studies of viral infections using multi-photon intravital microscopy (MP-IVM). Initially, the underlying principle of multi-photon microscopy is introduced and experimental challenges during microsurgical animal preparation and fluorescent labeling strategies for intravital imaging are discussed. I will further highlight recent studies that combine MP-IVM with optogenetic tools and transcriptional analysis as a powerful approach to extend the significance of in vivo imaging studies of viral pathogens.
Collapse
|
79
|
Benson RA, Garcon F, Recino A, Ferdinand JR, Clatworthy MR, Waldmann H, Brewer JM, Okkenhaug K, Cooke A, Garside P, Wållberg M. Non-Invasive Multiphoton Imaging of Islets Transplanted Into the Pinna of the NOD Mouse Ear Reveals the Immediate Effect of Anti-CD3 Treatment in Autoimmune Diabetes. Front Immunol 2018; 9:1006. [PMID: 29867981 PMCID: PMC5968092 DOI: 10.3389/fimmu.2018.01006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 04/23/2018] [Indexed: 12/16/2022] Open
Abstract
We present a novel and readily accessible method facilitating cellular time-resolved imaging of transplanted pancreatic islets. Grafting of islets to the mouse ear pinna allows non-invasive, in vivo longitudinal imaging of events in the islets and enables improved acquisition of experimental data and use of fewer experimental animals than is possible using invasive techniques, as the same mouse can be assessed for the presence of islet infiltrating cells before and after immune intervention. We have applied this method to investigating therapeutic protection of beta cells through the well-established use of anti-CD3 injection, and have acquired unprecedented data on the nature and rapidity of the effect on the islet infiltrating T cells. We demonstrate that infusion of anti-CD3 antibody leads to immediate effects on islet infiltrating T cells in islet grafts in the pinna of the ear, and causes them to increase their speed and displacement within 20 min of infusion. This technique overcomes several technical challenges associated with intravital imaging of pancreatic immune responses and facilitates routine study of beta islet cell development, differentiation, and function in health and disease.
Collapse
Affiliation(s)
- Robert A. Benson
- College of Medical, Veterinary & Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Fabien Garcon
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
| | - Asha Recino
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - John R. Ferdinand
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Menna R. Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Herman Waldmann
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - James M. Brewer
- College of Medical, Veterinary & Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Anne Cooke
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Paul Garside
- College of Medical, Veterinary & Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Maja Wållberg
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
80
|
Milo I, Blecher-Gonen R, Barnett-Itzhaki Z, Bar-Ziv R, Tal O, Gurevich I, Feferman T, Drexler I, Amit I, Bousso P, Shakhar G. The bone marrow is patrolled by NK cells that are primed and expand in response to systemic viral activation. Eur J Immunol 2018; 48:1137-1152. [PMID: 29624673 DOI: 10.1002/eji.201747378] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/30/2018] [Accepted: 03/27/2018] [Indexed: 12/11/2022]
Abstract
The bone marrow hosts NK cells whose distribution, motility and response to systemic immune challenge are poorly understood. At steady state, two-photon microscopy of the bone marrow in Ncr1gfp/+ mice captured motile NK cells interacting with dendritic cells. NK cells expressed markers and effector molecules of mature cells. Following poly (I:C) injection, RNA-Seq of NK cells revealed three phases of transcription featuring immune response genes followed by posttranscriptional processes and proliferation. Functionally, poly (I:C) promoted upregulation of granzyme B, enhanced cytotoxicity in vitro and in vivo, and, in the same individual cells, triggered proliferation. Two-photon imaging revealed that the proportion of sinusoidal NK cells decreased, while at the same time parenchymal NK cells accelerated, swelled and divided within the bone marrow. MVA viremia induced similar responses. Our findings demonstrate that the bone marrow is patrolled by mature NK cells that rapidly proliferate in response to systemic viral challenge while maintaining their effector functions.
Collapse
Affiliation(s)
- Idan Milo
- Department of Immunology, the Weizmann Institute of Science, Rehovot, Israel.,Institut Pasteur, Dynamics of Immune Responses Unit, Equipe Labéllisée Ligue Contre le Cancer, Institut Pasteur, Paris, France
| | | | | | - Raz Bar-Ziv
- Department of Immunology, the Weizmann Institute of Science, Rehovot, Israel
| | - Orna Tal
- Department of Immunology, the Weizmann Institute of Science, Rehovot, Israel
| | - Irina Gurevich
- Department of Immunology, the Weizmann Institute of Science, Rehovot, Israel
| | - Tali Feferman
- Department of Immunology, the Weizmann Institute of Science, Rehovot, Israel
| | - Ingo Drexler
- Institute for Virology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ido Amit
- Department of Immunology, the Weizmann Institute of Science, Rehovot, Israel
| | - Philippe Bousso
- Institut Pasteur, Dynamics of Immune Responses Unit, Equipe Labéllisée Ligue Contre le Cancer, Institut Pasteur, Paris, France
| | - Guy Shakhar
- Department of Immunology, the Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
81
|
Tsukahara T, Nakamura SI, Romero-Pèrez GA, Ohwaki M, Yanagisawa T, Kan T. Stimulation of murine cell-mediated immunity by dietary administration of a cell preparation of Enterococcus faecalis strain KH-2 and its possible activity against tumour development in mice. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2018; 37:49-57. [PMID: 30094120 PMCID: PMC6081610 DOI: 10.12938/bmfh.17-021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/09/2018] [Indexed: 01/09/2023]
Abstract
It is well known that dietary lactic acid bacteria (LAB) stimulate cell-mediated immunity such as natural killer (NK) activity in mice. Here, we aimed to assay the immunomodulatory effects of a cell preparation of Enterococcus faecalis strain KH-2 (CPEF). We further evaluated the possibility of antitumour activity caused by CPEF administration, because NK cells actively participate in the prevention of tumour formation. NK cell activity and gene expression of IFN-γ and Perforin 1, which were induced most likely by a synergetic action of their cytotoxic activity, were higher in splenocytes of CPEF-administered mice than they were in control mice. Moreover, unlike those of control mice, the splenocytes of CPEF-administered mice had significantly higher CD28+CD69+/CD4+ and CD28+CD69+/CD8+ ratios that resulted in a survival rate with a tendency toward improvement after 47 days of CPEF administration (p=0.1) in Meth-A fibrosarcoma-bearing mice. In conclusion, we showed that CPEF might be effective in treating Meth-A fibrosarcoma in mice, as it helped increase their survival rate via stimulation of an immune response in splenocytes, which involved systemic cellular immunity processes such as cytotoxic activity, and active T cells.
Collapse
Affiliation(s)
- Takamitsu Tsukahara
- Kyoto Institute of Nutrition and Pathology, 7-2 Furuikedani, Ujitawara-cho, Tsuzuki-gun, Kyoto 610-0231, Japan
| | - Shin-Ichi Nakamura
- Kyoto Institute of Nutrition and Pathology, 7-2 Furuikedani, Ujitawara-cho, Tsuzuki-gun, Kyoto 610-0231, Japan
| | - Gustavo A Romero-Pèrez
- Kyoto Institute of Nutrition and Pathology, 7-2 Furuikedani, Ujitawara-cho, Tsuzuki-gun, Kyoto 610-0231, Japan
| | - Makoto Ohwaki
- Non-Profit Organisation, The Japanese Association of Clinical Research on Supplements, 1-9-24 Shihogi, Hidaka-shi, Saitama 350-1248, Japan
| | - Takaharu Yanagisawa
- Broma Laboratory Ltd., 1-26 Kandasuda-cho, Chiyoda-ku, Tokyo 101-0041, Japan
| | - Tatsuhiko Kan
- Bio-Lab Co., Ltd., 2-1-3 Komagawa, Hidaka-shi, Saitama 350-1249, Japan
| |
Collapse
|
82
|
Imaging of cytotoxic antiviral immunity while considering the 3R principle of animal research. J Mol Med (Berl) 2018; 96:349-360. [PMID: 29460050 DOI: 10.1007/s00109-018-1628-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/01/2018] [Accepted: 02/05/2018] [Indexed: 10/18/2022]
Abstract
Adoptive cell transfer approaches for antigen-specific CD8+ T cells are used widely to study their effector potential during infections or cancer. However, contemporary methodological adaptations regarding transferred cell numbers, advanced imaging, and the 3R principle of animal research have been largely omitted. Here, we introduce an improved cell transfer method that reduces the number of donor animals substantially and fulfills the requirements for intravital imaging under physiological conditions. For this, we analyzed the well-established Friend retrovirus (FV) mouse model. Donor mice that expressed a FV-specific T cell receptor (TCRtg) and the fluorescent protein tdTomato were used as source of antigen-specific CD8+ T cells. Only a few drops of peripheral blood were sufficient to isolate ~ 150,000 naive reporter cells from which 1000 were adoptively transferred into recently FV-infected recipients. The cells became activated and functional and expanded strongly in the spleen and bone marrow within 10 days post infection. Transferred CD8+ T cells participated in the antiviral host response within a natural range and developed an effector phenotype indistinguishable from endogenous effector CD8+ T cells. Additionally, the generated reporter cell frequency allowed single cell visualization and tracking of a physiological antiretroviral CD8+ T cell response by intravital two-photon microscopy. Highly reproducible results were obtained in independent experiments by reusing the same donors repetitively for multiple transfers. Our approach allows a strong reduction of experimental animals required for studies on antigen-specific CD8+ T cell function and should be applicable to other transfer models. KEY MESSAGES TCRtg CD8+ T cells are obtained repetitively from the blood samples of single donors. One thousand transferred TCRtg CD8+ T cells get activated, are functional, and proliferate. Several adoptive cell transfers from the same donor show reproducible results. One thousand transferred cells take part in the FV immune response without modifying it. Use of fluorescent transfer cells allows in vivo imaging and single cell tracking.
Collapse
|
83
|
Zhou J, Bethune MT, Malkova N, Sutherland AM, Comin-Anduix B, Su Y, Baltimore D, Ribas A, Heath JR. A kinetic investigation of interacting, stimulated T cells identifies conditions for rapid functional enhancement, minimal phenotype differentiation, and improved adoptive cell transfer tumor eradication. PLoS One 2018; 13:e0191634. [PMID: 29360859 PMCID: PMC5779691 DOI: 10.1371/journal.pone.0191634] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/09/2018] [Indexed: 12/04/2022] Open
Abstract
For adoptive cell transfer (ACT) immunotherapy of tumor-reactive T cells, an effective therapeutic outcome depends upon cell dose, cell expansion in vivo through a minimally differentiated phenotype, long term persistence, and strong cytolytic effector function. An incomplete understanding of the biological coupling between T cell expansion, differentiation, and response to stimulation hinders the co-optimization of these factors. We report on a biophysical investigation of how the short-term kinetics of T cell functional activation, through molecular stimulation and cell-cell interactions, competes with phenotype differentiation. T cells receive molecular stimulation for a few minutes to a few hours in bulk culture. Following this priming period, the cells are then analyzed at the transcriptional level, or isolated as single cells, with continuing molecular stimulation, within microchambers for analysis via 11-plex secreted protein assays. We resolve a rapid feedback mechanism, promoted by T cell—T cell contact interactions, which strongly amplifies T cell functional performance while yielding only minimal phenotype differentiation. When tested in mouse models of ACT, optimally primed T cells lead to complete tumor eradication. A similar kinetic process is identified in CD8+ and CD4+ T cells collected from a patient with metastatic melanoma.
Collapse
Affiliation(s)
- Jing Zhou
- NanoSystems Biology Cancer Center, California Institute of Technology, Pasadena, California, United States of America.,Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Michael T Bethune
- NanoSystems Biology Cancer Center, California Institute of Technology, Pasadena, California, United States of America.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Natalia Malkova
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Alexander M Sutherland
- NanoSystems Biology Cancer Center, California Institute of Technology, Pasadena, California, United States of America.,Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Begonya Comin-Anduix
- David Geffen School of Medicine, the Johnson Comprehensive Cancer Center, University of California, Los Angeles, California, United States of America
| | - Yapeng Su
- NanoSystems Biology Cancer Center, California Institute of Technology, Pasadena, California, United States of America.,Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - David Baltimore
- NanoSystems Biology Cancer Center, California Institute of Technology, Pasadena, California, United States of America.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Antoni Ribas
- NanoSystems Biology Cancer Center, California Institute of Technology, Pasadena, California, United States of America.,David Geffen School of Medicine, the Johnson Comprehensive Cancer Center, University of California, Los Angeles, California, United States of America
| | - James R Heath
- NanoSystems Biology Cancer Center, California Institute of Technology, Pasadena, California, United States of America.,Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
| |
Collapse
|
84
|
Garetto S, Sardi C, Martini E, Roselli G, Morone D, Angioni R, Cianciotti BC, Trovato AE, Franchina DG, Castino GF, Vignali D, Erreni M, Marchesi F, Rumio C, Kallikourdis M. Tailored chemokine receptor modification improves homing of adoptive therapy T cells in a spontaneous tumor model. Oncotarget 2018; 7:43010-43026. [PMID: 27177227 PMCID: PMC5190004 DOI: 10.18632/oncotarget.9280] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 04/27/2016] [Indexed: 01/19/2023] Open
Abstract
In recent years, tumor Adoptive Cell Therapy (ACT), using administration of ex vivo-enhanced T cells from the cancer patient, has become a promising therapeutic strategy. However, efficient homing of the anti-tumoral T cells to the tumor or metastatic site still remains a substantial hurdle. Yet the tumor site itself attracts both tumor-promoting and anti-tumoral immune cell populations through the secretion of chemokines. We attempted to identify these chemokines in a model of spontaneous metastasis, in order to “hijack” their function by expressing matching chemokine receptors on the cytotoxic T cells used in ACT, thus allowing us to enhance the recruitment of these therapeutic cells. Here we show that this enabled the modified T cells to preferentially home into spontaneous lymph node metastases in the TRAMP model, as well as in an inducible tumor model, E.G7-OVA. Due to the improved homing, the modified CD8+ T cells displayed an enhanced in vivo protective effect, as seen by a significant delay in E.G7-OVA tumor growth. These results offer a proof of principle for the tailored application of chemokine receptor modification as a means of improving T cell homing to the target tumor, thus enhancing ACT efficacy. Surprisingly, we also uncover that the formation of the peri-tumoral fibrotic capsule, which has been shown to impede T cell access to tumor, is partially dependent on host T cell presence. This finding, which would be impossible to observe in immunodeficient model studies, highlights possible conflicting roles that T cells may play in a therapeutic context.
Collapse
Affiliation(s)
- Stefano Garetto
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Rozzano (Milano), Italy
| | - Claudia Sardi
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Rozzano (Milano), Italy
| | - Elisa Martini
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Rozzano (Milano), Italy
| | - Giuliana Roselli
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Rozzano (Milano), Italy
| | - Diego Morone
- Humanitas Clinical and Research Center, Rozzano (Milano), Italy
| | - Roberta Angioni
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Rozzano (Milano), Italy
| | | | - Anna Elisa Trovato
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Rozzano (Milano), Italy
| | | | | | - Debora Vignali
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Rozzano (Milano), Italy
| | - Marco Erreni
- Laboratory of Cellular Immunology, Humanitas Clinical and Research Center, Rozzano (Milano), Italy
| | - Federica Marchesi
- Laboratory of Cellular Immunology, Humanitas Clinical and Research Center, Rozzano (Milano), Italy.,Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Cristiano Rumio
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Marinos Kallikourdis
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Rozzano (Milano), Italy.,Humanitas University, Rozzano (Milano), Italy
| |
Collapse
|
85
|
Denton AE, Roberts EW, Fearon DT. Stromal Cells in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:99-114. [PMID: 30155624 DOI: 10.1007/978-3-319-78127-3_6] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The tumor microenvironment comprises a mass of heterogeneous cell types, including immune cells, endothelial cells, and fibroblasts, alongside cancer cells. It is increasingly becoming clear that the development of this support niche is critical to the continued uncontrolled growth of the cancer. The tumor microenvironment contributes to the maintenance of cancer stemness and also directly promotes angiogenesis, invasion, metastasis, and chronic inflammation. In this chapter, we describe on the role of fibroblasts, specifically termed cancer-associated fibroblasts (CAFs), in the promotion and maintenance of cancers. CAFs have a multitude of effects on the growth and maintenance of cancer, and here we focus on their roles in modulating immune cells and responses; CAFs both inhibit immune cell access to the tumor microenvironment and inhibit their functions within the tumor. Finally, we describe the potential modulation of CAF function as an adjunct to bolster the effectiveness of cancer immunotherapies.
Collapse
Affiliation(s)
- Alice E Denton
- Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK.
| | - Edward W Roberts
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Douglas T Fearon
- Cold Spring Harbor Laboratory, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
86
|
Owyong M, Hosseini-Nassab N, Efe G, Honkala A, van den Bijgaart RJE, Plaks V, Smith BR. Cancer Immunotherapy Getting Brainy: Visualizing the Distinctive CNS Metastatic Niche to Illuminate Therapeutic Resistance. Drug Resist Updat 2017; 33-35:23-35. [PMID: 29145972 DOI: 10.1016/j.drup.2017.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The advent of cancer immunotherapy (CIT) and its success in treating primary and metastatic cancer may offer substantially improved outcomes for patients. Despite recent advancements, many malignancies remain resistant to CIT, among which are brain metastases, a particularly virulent disease with no apparent cure. The immunologically unique niche of the brain has prompted compelling new questions in immuno-oncology such as the effects of tissue-specific differences in immune response, heterogeneity between primary tumors and distant metastases, and the role of spatiotemporal dynamics in shaping an effective anti-tumor immune response. Current methods to examine the immunobiology of metastases in the brain are constrained by tissue processing methods that limit spatial data collection, omit dynamic information, and cannot recapitulate the heterogeneity of the tumor microenvironment. In the current review, we describe how high-resolution, live imaging tools, particularly intravital microscopy (IVM), are instrumental in answering these questions. IVM of pre-clinical cancer models enables short- and long-term observations of critical immunobiology and metastatic growth phenomena to potentially generate revolutionary insights into the spatiotemporal dynamics of brain metastasis, interactions of CIT with immune elements therein, and influence of chemo- and radiotherapy. We describe the utility of IVM to study brain metastasis in mice by tracking the migration and growth of fluorescently-labeled cells, including cancer cells and immune subsets, while monitoring the physical environment within optical windows using imaging dyes and other signal generation mechanisms to illuminate angiogenesis, hypoxia, and/or CIT drug expression within the metastatic niche. Our review summarizes the current knowledge regarding brain metastases and the immune milieu, presents the current status of CIT and its prospects in targeting brain metastases to circumvent therapeutic resistance, and proposes avenues to utilize IVM to study CIT drug delivery and therapeutic efficacy in preclinical models that will ultimately facilitate novel drug discovery and innovative combination therapies.
Collapse
Affiliation(s)
- Mark Owyong
- Department of Anatomy, University of California, San Francisco, CA 94143-0452, USA
| | | | - Gizem Efe
- Department of Anatomy, University of California, San Francisco, CA 94143-0452, USA
| | - Alexander Honkala
- Department of Radiology, Stanford University, Stanford, CA 94306, USA
| | - Renske J E van den Bijgaart
- Department of Radiation Oncology, Radiotherapy and Oncoimmunology Laboratory, Radboudumc, Geert Grooteplein Zuid 32, 6525, GA, Nijmegen, The Netherlands
| | - Vicki Plaks
- Department of Orofacial Sciences, University of California, San Francisco, CA 94143, USA.
| | | |
Collapse
|
87
|
Kather JN, Poleszczuk J, Suarez-Carmona M, Krisam J, Charoentong P, Valous NA, Weis CA, Tavernar L, Leiss F, Herpel E, Klupp F, Ulrich A, Schneider M, Marx A, Jäger D, Halama N. In Silico Modeling of Immunotherapy and Stroma-Targeting Therapies in Human Colorectal Cancer. Cancer Res 2017; 77:6442-6452. [PMID: 28923860 DOI: 10.1158/0008-5472.can-17-2006] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/21/2017] [Accepted: 09/13/2017] [Indexed: 12/29/2022]
Abstract
Despite the fact that the local immunological microenvironment shapes the prognosis of colorectal cancer, immunotherapy has shown no benefit for the vast majority of colorectal cancer patients. A better understanding of the complex immunological interplay within the microenvironment is required. In this study, we utilized wet lab migration experiments and quantitative histological data of human colorectal cancer tissue samples (n = 20) including tumor cells, lymphocytes, stroma, and necrosis to generate a multiagent spatial model. The resulting data accurately reflected a wide range of situations of successful and failed immune surveillance. Validation of simulated tissue outcomes on an independent set of human colorectal cancer specimens (n = 37) revealed the model recapitulated the spatial layout typically found in human tumors. Stroma slowed down tumor growth in a lymphocyte-deprived environment but promoted immune escape in a lymphocyte-enriched environment. A subgroup of tumors with less stroma and high numbers of immune cells showed high rates of tumor control. These findings were validated using data from colorectal cancer patients (n = 261). Low-density stroma and high lymphocyte levels showed increased overall survival (hazard ratio 0.322, P = 0.0219) as compared with high stroma and high lymphocyte levels. To guide immunotherapy in colorectal cancer, simulation of immunotherapy in preestablished tumors showed that a complex landscape with optimal stroma permeabilization and immune cell activation is able to markedly increase therapy response in silico These results can help guide the rational design of complex therapeutic interventions, which target the colorectal cancer microenvironment. Cancer Res; 77(22); 6442-52. ©2017 AACR.
Collapse
Affiliation(s)
- Jakob Nikolas Kather
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Poleszczuk
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Meggy Suarez-Carmona
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Johannes Krisam
- Institute of Medical Biometry and Informatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Pornpimol Charoentong
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nektarios A Valous
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cleo-Aron Weis
- Department of Pathology, University Medical Center Mannheim, Mannheim, Germany
| | - Luca Tavernar
- Institute of Pathology, Heidelberg University, Heidelberg, Germany.,Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, Germany
| | | | - Esther Herpel
- Institute of Pathology, Heidelberg University, Heidelberg, Germany.,Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, Germany
| | - Fee Klupp
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexander Marx
- Department of Pathology, University Medical Center Mannheim, Mannheim, Germany
| | - Dirk Jäger
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Niels Halama
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany. .,German Cancer Consortium (DKTK), Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
88
|
An X, Sendra VG, Liadi I, Ramesh B, Romain G, Haymaker C, Martinez-Paniagua M, Lu Y, Radvanyi LG, Roysam B, Varadarajan N. Single-cell profiling of dynamic cytokine secretion and the phenotype of immune cells. PLoS One 2017; 12:e0181904. [PMID: 28837583 PMCID: PMC5570329 DOI: 10.1371/journal.pone.0181904] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/10/2017] [Indexed: 01/05/2023] Open
Abstract
Natural killer (NK) cells are a highly heterogeneous population of innate lymphocytes that constitute our first line of defense against several types of tumors and microbial infections. Understanding the heterogeneity of these lymphocytes requires the ability to integrate their underlying phenotype with dynamic functional behaviors. We have developed and validated a single-cell methodology that integrates cellular phenotyping and dynamic cytokine secretion based on nanowell arrays and bead-based molecular biosensors. We demonstrate the robust passivation of the polydimethylsiloxane (PDMS)-based nanowells arrays with polyethylene glycol (PEG) and validated our assay by comparison to enzyme-linked immunospot (ELISPOT) assays. We used numerical simulations to optimize the molecular density of antibodies on the surface of the beads as a function of the capture efficiency of cytokines within an open-well system. Analysis of hundreds of individual human peripheral blood NK cells profiled ex vivo revealed that CD56dimCD16+ NK cells are immediate secretors of interferon gamma (IFN-γ) upon activation by phorbol 12-myristate 13-acetate (PMA) and ionomycin (< 3 h), and that there was no evidence of cooperation between NK cells leading to either synergistic activation or faster IFN-γ secretion. Furthermore, we observed that both the amount and rate of IFN-γ secretion from individual NK cells were donor-dependent. Collectively, these results establish our methodology as an investigational tool for combining phenotyping and real-time protein secretion of individual cells in a high-throughput manner.
Collapse
Affiliation(s)
- Xingyue An
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, United States of America
| | - Victor G. Sendra
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, United States of America
| | - Ivan Liadi
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, United States of America
| | - Balakrishnan Ramesh
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, United States of America
| | - Gabrielle Romain
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, United States of America
| | - Cara Haymaker
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Melisa Martinez-Paniagua
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, United States of America
| | - Yanbin Lu
- Department of Electrical and Computer Engineering, University of Houston, Houston, Texas, United States of America
| | - Laszlo G. Radvanyi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Badrinath Roysam
- Department of Electrical and Computer Engineering, University of Houston, Houston, Texas, United States of America
| | - Navin Varadarajan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
89
|
Wang M, Yu Y, Shao J, Heng BC, Ye H. Engineering synthetic optogenetic networks for biomedical applications. QUANTITATIVE BIOLOGY 2017. [DOI: 10.1007/s40484-017-0105-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
90
|
Halle S, Halle O, Förster R. Mechanisms and Dynamics of T Cell-Mediated Cytotoxicity In Vivo. Trends Immunol 2017; 38:432-443. [PMID: 28499492 DOI: 10.1016/j.it.2017.04.002] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 02/06/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) are critical in the elimination of infected or malignant cells and are emerging as a major therapeutic target. How CTLs recognize and kill harmful cells has been characterized in vitro but little is known about these processes in the living organism. Here we review recent insights into CTL-mediated killing with an emphasis on in vivo CTL biology. Specifically, we focus on the possible rate-limiting steps determining the efficiency of CTL-mediated killing. We also highlight the need for cell-based datasets that permit the quantification of CTL dynamics, including CTL location, migration, and killing rates. A better understanding of these factors is required to predict protective CD8 T cell immunity in vivo and to design optimized vaccination protocols.
Collapse
Affiliation(s)
- Stephan Halle
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany.
| | - Olga Halle
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
91
|
Suijkerbuijk SJE, van Rheenen J. From good to bad: Intravital imaging of the hijack of physiological processes by cancer cells. Dev Biol 2017; 428:328-337. [PMID: 28473106 DOI: 10.1016/j.ydbio.2017.04.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 12/23/2022]
Abstract
Homeostasis of tissues is tightly regulated at the cellular, tissue and organismal level. Interestingly, tumor cells have found ways to hijack many of these physiological processes at all the different levels. Here we review how intravital microscopy techniques have provided new insights into our understanding of tissue homeostasis and cancer progression. In addition, we highlight the different strategies that tumor cells have adopted to use these physiological processes for their own benefit. We describe how visualization of these dynamic processes in living mice has broadened to our view on cancer initiation and progression.
Collapse
Affiliation(s)
- Saskia J E Suijkerbuijk
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, 3584 CG Utrecht, The Netherlands
| | - Jacco van Rheenen
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, 3584 CG Utrecht, The Netherlands.
| |
Collapse
|
92
|
Torcellan T, Stolp J, Chtanova T. In Vivo Imaging Sheds Light on Immune Cell Migration and Function in Cancer. Front Immunol 2017; 8:309. [PMID: 28382036 PMCID: PMC5360706 DOI: 10.3389/fimmu.2017.00309] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/06/2017] [Indexed: 01/04/2023] Open
Abstract
There is ample evidence for both beneficial and harmful involvement of the immune system in tumor development and spread. Immune cell recruitment to tumors is essential not only for the success of anticancer immune therapies but also for tumor-induced immune suppression. Now that immune-based therapies are playing an increasingly important role in treatment of solid tumors such as metastatic melanomas, precise analysis of the in vivo contributions of different leukocyte subsets in tumor immunity has become an even greater priority. Recently, this goal has been markedly facilitated by the use of intravital microscopy, which has enabled us to visualize the dynamic interactions between cells of the immune system and tumor targets in the context of the tumor microenvironment. For example, intravital imaging techniques have shed new light on T cell infiltration of tumors, the mechanisms of cancer cell killing, and how myeloid cells contribute to tumor tolerance and spread. This mini-review summarizes the recent advances made to our understanding of the roles of innate and adaptive immune cells in cancer based on the use of these in vivo imaging approaches.
Collapse
Affiliation(s)
- Tommaso Torcellan
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Jessica Stolp
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Tatyana Chtanova
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
93
|
Gadhamsetty S, Marée AFM, de Boer RJ, Beltman JB. Tissue Dimensionality Influences the Functional Response of Cytotoxic T Lymphocyte-Mediated Killing of Targets. Front Immunol 2017; 7:668. [PMID: 28123385 PMCID: PMC5225319 DOI: 10.3389/fimmu.2016.00668] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/19/2016] [Indexed: 11/13/2022] Open
Abstract
Cytotoxic T lymphocyte (CTL)-mediated killing of virus infections and tumors occurs over a wide range of conditions. The spatial environments in which CTLs encounter target cells vary from narrow vessels, to two-dimensional epithelial tissues, to densely populated 3-dimensional (3D) T cell areas within lymphoid tissues. How such spatial environments alter the functional response of CTL-mediated killing, i.e., how the killing efficiency depends on cell densities, is unclear. In this study, we perform cellular Potts model simulations in different spatial configurations to investigate how the dimensionality of the space affects the functional response of CTL-mediated killing. Irrespective of the spatial configuration, the function with separate saturation constants for CTL and for target cell densities that we previously proposed can in all cases describe the response, demonstrating its generality. However, the tissue dimensionality determines at which cell densities the killing rate starts to saturate. We show that saturation in a fully 3D environment is stronger than in a "flat" 3D environment, which is largely due to accompanying differences in the CTL-target encounter rates.
Collapse
Affiliation(s)
| | - Athanasius F M Marée
- Department of Computational and Systems Biology, John Innes Centre , Norwich , UK
| | - Rob J de Boer
- Theoretical Biology, Utrecht University , Utrecht , Netherlands
| | - Joost B Beltman
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University , Leiden , Netherlands
| |
Collapse
|
94
|
Kavunja HW, Lang S, Sungsuwan S, Yin Z, Huang X. Delivery of foreign cytotoxic T lymphocyte epitopes to tumor tissues for effective antitumor immunotherapy against pre-established solid tumors in mice. Cancer Immunol Immunother 2016; 66:451-460. [PMID: 28011995 DOI: 10.1007/s00262-016-1948-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 12/18/2016] [Indexed: 11/26/2022]
Abstract
Cytotoxic T lymphocyte (CTL) can have remarkable abilities to kill tumor cells. However, the establishment of successful CTL-based anticancer therapy has met with many challenges. Within tumor cells, there exist subpopulations with low or no expression of the targeted antigen (termed as antigen-loss variants). In addition, tumor cells can downregulate the levels of major histocompatibility complex class I (MHC-I) molecules on cell surface due to immune pressure. As a result, some tumor cells can escape the immune pressure bestowed by CTLs, resulting in treatment failure. To address these difficulties, a new approach is developed to deliver foreign high-affinity CTL epitopes to tumor tissues utilizing pH-responsive "smart" microparticles (MPs). These MPs could encapsulate CTL peptide epitope, release the peptide under acidic condition encountered in tumor tissues and enhance CTL activation. Mice bearing pre-established tumor as "antigen-loss variant" solid tumor models were administered intratumorally with MPs containing the CTL peptide, which showed 100% survival following the treatment. In contrast, all control mice died from tumor. Significant protection from tumor-induced death was also observed with systemic administration of CTL peptide-MPs. The therapeutic efficacy can be attributed to enhanced delivery of the epitope to tumor tissues, presentation of the epitope by tumor cells as well as tumor stromal cells and/or generation of epitope-specific CTLs by the peptide-containing MPs. These findings offer a promising new direction for treating established solid tumor using CTL therapy.
Collapse
Affiliation(s)
- Herbert W Kavunja
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
| | - Shuyao Lang
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
| | - Suttipun Sungsuwan
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
| | - Zhaojun Yin
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA.
| |
Collapse
|
95
|
Qi S, Li H, Lu L, Qi Z, Liu L, Chen L, Shen G, Fu L, Luo Q, Zhang Z. Long-term intravital imaging of the multicolor-coded tumor microenvironment during combination immunotherapy. eLife 2016; 5. [PMID: 27855783 PMCID: PMC5173323 DOI: 10.7554/elife.14756] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 10/17/2016] [Indexed: 12/22/2022] Open
Abstract
The combined-immunotherapy of adoptive cell therapy (ACT) and cyclophosphamide (CTX) is one of the most efficient treatments for melanoma patients. However, no synergistic effects of CTX and ACT on the spatio-temporal dynamics of immunocytes in vivo have been described. Here, we visualized key cell events in immunotherapy-elicited immunoreactions in a multicolor-coded tumor microenvironment, and then established an optimal strategy of metronomic combined-immunotherapy to enhance anti-tumor efficacy. Intravital imaging data indicated that regulatory T cells formed an 'immunosuppressive ring' around a solid tumor. The CTX-ACT combined-treatment elicited synergistic immunoreactions in tumor areas, which included relieving the immune suppression, triggering the transient activation of endogenous tumor-infiltrating immunocytes, increasing the accumulation of adoptive cytotoxic T lymphocytes, and accelerating the infiltration of dendritic cells. These insights into the spatio-temporal dynamics of immunocytes are beneficial for optimizing immunotherapy and provide new approaches for elucidating the mechanisms underlying the involvement of immunocytes in cancer immunotherapy. DOI:http://dx.doi.org/10.7554/eLife.14756.001 Melanoma is a form of skin cancer that is particularly difficult to treat. A new approach that has shown a lot of promise in treating many different cancers, including melanoma, is called “immunotherapy”. This technique harnesses the immune system – the body’s natural defences that help to protect against infections and disease – to combat cancer. One powerful type of immunotherapy involves injecting patients with cells called lymphocytes, which form part of the immune system. This is known as adoptive cell therapy and can activate the immune system to fight cancer, helping to shrink tumors. This treatment can be made even more powerful by combining it with a drug called cyclophosphamide and this combination, known as CTX-ACT, is currently one of the most efficient treatments for melanoma. Yet, little information is available to indicate why this treatment is so effective. Using mice implanted with melanoma cells, Qi, Li et al. sought to understand how CTX-ACT treatment works, with the goal of optimising it to increase its success. The results showed that a protective barrier of immune cells that suppresses the anti-tumor immune response – called an “immunosuppressive ring” – surrounds untreated tumors. CTX-ACT treatment can breakdown these rings, helping to reactivate the anti-tumor immune reaction in the tumors. This allows both the injected and mouse’s own immune cells to move into the tumor and destroy cancer cells. Qi, Li et al. used their findings to optimise treatment and succeeded in controlling tumor growth in the mice for several weeks. These new insights could be used to improve current immunotherapies, and offer new approaches for investigating the involvement of immune cells in the treatment of a wide range of different cancers. DOI:http://dx.doi.org/10.7554/eLife.14756.002
Collapse
Affiliation(s)
- Shuhong Qi
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Lisen Lu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Zhongyang Qi
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China.,Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanxin Shen
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Fu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihong Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
96
|
Sun TY, Haberman AM, Greco V. Preclinical Advances with Multiphoton Microscopy in Live Imaging of Skin Cancers. J Invest Dermatol 2016; 137:282-287. [PMID: 27847119 DOI: 10.1016/j.jid.2016.08.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 08/09/2016] [Accepted: 08/24/2016] [Indexed: 01/13/2023]
Abstract
Conventional, static analyses have historically been the bedrock and tool of choice for the study of skin cancers. Over the past several years, in vivo imaging of tumors using multiphoton microscopy has emerged as a powerful preclinical tool for revealing detailed cellular behaviors from the earliest moments of tumor development to the final steps of metastasis. Multiphoton microscopy allows for deep tissue penetration with relatively minor phototoxicity, rendering it an effective tool for the long-term observation of tumor evolution. This review highlights some of the recent preclinical insights gained using multiphoton microscopy and suggests future advances that could enhance its power in revealing the mysteries of skin tumor biology.
Collapse
Affiliation(s)
- Thomas Yang Sun
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA.
| | - Ann M Haberman
- Departments of Immunobiology and Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Valentina Greco
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA; Departments of Dermatology and Cell Biology, Yale Stem Cell Center, Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
97
|
Hsu FT, Chen TC, Chuang HY, Chang YF, Hwang JJ. Enhancement of adoptive T cell transfer with single low dose pretreatment of doxorubicin or paclitaxel in mice. Oncotarget 2016; 6:44134-50. [PMID: 26683520 PMCID: PMC4792547 DOI: 10.18632/oncotarget.6628] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 11/25/2015] [Indexed: 01/17/2023] Open
Abstract
Ex vivo expansion of CD8+ T-cells has been a hindrance for the success of adoptive T cell transfer in clinic. Currently, preconditioning with chemotherapy is used to modulate the patient immunity before ACT, however, the tumor microenvironment beneficial for transferring T cells may also be damaged. Here preconditioning with single low dose of doxorubicin or paclitaxel combined with fewer CD8+ T-cells was investigated to verify whether the same therapeutic efficacy of ACT could be achieved. An E.G7/OT1 animal model that involved adoptive transfer of OVA-specific CD8+ T-cells transduced with a granzyme B promoter-driven firefly luciferase and tomato fluorescent fusion reporter gene was used to evaluate this strategy. The result showed that CD8+ T-cells were activated and sustained longer in mice pretreated with one low-dose Dox or Tax. Enhanced therapeutic efficacy was found in Dox or Tax combined with 2x106 CD8+ T-cells and achieved the same level of tumor growth inhibition as that of 5x106 CD8+ T-cells group. Notably, reduced numbers of Tregs and myeloid derived suppressor cells were shown in combination groups. By contrast, the number of tumor-infiltrating cytotoxic T lymphocytes and IL-12 were increased. The NF-κB activity and immunosuppressive factors such as TGF-β, IDO, CCL2, VEGF, CCL22, COX-2 and IL-10 were suppressed. This study demonstrates that preconditioning with single low dose Dox or Tax and combined with two fifth of the original CD8+ T-cells could improve the tumor microenvironment via suppression of NF-κB and its related immunosuppressors, and activate more CD8+ T-cells which also stay longer.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan.,Translational Imaging Research Center, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Chun Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Hui-Yen Chuang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ya-Fang Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Jeng-Jong Hwang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Biophotonics and Molecular Imaging Research Center (BMIRC), National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
98
|
Vaccine-induced tumor regression requires a dynamic cooperation between T cells and myeloid cells at the tumor site. Oncotarget 2016; 6:27832-46. [PMID: 26337837 PMCID: PMC4695029 DOI: 10.18632/oncotarget.4940] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/29/2015] [Indexed: 12/31/2022] Open
Abstract
Most cancer immunotherapies under present investigation are based on the belief that cytotoxic T cells are the most important anti-tumoral immune cells, whereas intra-tumoral macrophages would rather play a pro-tumoral role. We have challenged this antagonistic point of view and searched for collaborative contributions by tumor-infiltrating T cells and macrophages, reminiscent of those observed in anti-infectious responses. We demonstrate that, in a model of therapeutic vaccination, cooperation between myeloid cells and T cells is indeed required for tumor rejection. Vaccination elicited an early rise of CD11b+ myeloid cells that preceded and conditioned the intra-tumoral accumulation of CD8+ T cells. Conversely, CD8+ T cells and IFNγ production activated myeloid cells were required for tumor regression. A 4-fold reduction of CD8+ T cell infiltrate in CXCR3KO mice did not prevent tumor regression, whereas a reduction of tumor-infiltrating myeloid cells significantly interfered with vaccine efficiency. We show that macrophages from regressing tumors can kill tumor cells in two ways: phagocytosis and TNFα release. Altogether, our data suggest new strategies to improve the efficiency of cancer immunotherapies, by promoting intra-tumoral cooperation between macrophages and T cells.
Collapse
|
99
|
Yang F, Liu S, Liu X, Liu L, Luo M, Qi S, Xu G, Qiao S, Lv X, Li X, Fu L, Luo Q, Zhang Z. In Vivo Visualization of Tumor Antigen-containing Microparticles Generated in Fluorescent-protein-elicited Immunity. Theranostics 2016; 6:1453-66. [PMID: 27375792 PMCID: PMC4924512 DOI: 10.7150/thno.14145] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 05/05/2016] [Indexed: 02/07/2023] Open
Abstract
In vivo optical spatio-temporal imaging of the tumor microenvironment is useful to explain how tumor immunotherapies work. However, the lack of fluorescent antigens with strong immunogenicity makes it difficult to study the dynamics of how tumors are eliminated by any given immune response. Here, we develop an effective fluorescent model antigen based on the tetrameric far-red fluorescent protein KatushkaS158A (tfRFP), which elicits both humoral and cellular immunity. We use this fluorescent antigen to visualize the dynamic behavior of immunocytes as they attack and selectively eliminate tfRFP-expressing tumors in vivo; swarms of immunocytes rush toward tumors with high motility, clusters of immunocytes form quickly, and numerous antigen-antibody complexes in the form of tfRFP(+) microparticles are generated in the tumor areas and ingested by macrophages in the tumor microenvironment. Therefore, tfRFP, as both a model antigen and fluorescent reporter, is a useful tool to visualize specific immune responses in vivo.
Collapse
Affiliation(s)
- Fei Yang
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shun Liu
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiuli Liu
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lei Liu
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Meijie Luo
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shuhong Qi
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Guoqiang Xu
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Sha Qiao
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaohua Lv
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangning Li
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ling Fu
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qingming Luo
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhihong Zhang
- 1. Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- 2. MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
100
|
Real-time intravital imaging of pH variation associated with osteoclast activity. Nat Chem Biol 2016; 12:579-85. [PMID: 27272564 DOI: 10.1038/nchembio.2096] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 03/24/2016] [Indexed: 11/08/2022]
Abstract
Intravital imaging by two-photon excitation microscopy (TPEM) has been widely used to visualize cell functions. However, small molecular probes (SMPs), commonly used for cell imaging, cannot be simply applied to intravital imaging because of the challenge of delivering them into target tissues, as well as their undesirable physicochemical properties for TPEM imaging. Here, we designed and developed a functional SMP with an active-targeting moiety, higher photostability, and a fluorescence switch and then imaged target cell activity by injecting the SMP into living mice. The combination of the rationally designed SMP with a fluorescent protein as a reporter of cell localization enabled quantitation of osteoclast activity and time-lapse imaging of its in vivo function associated with changes in cell deformation and membrane fluctuations. Real-time imaging revealed heterogenic behaviors of osteoclasts in vivo and provided insights into the mechanism of bone resorption.
Collapse
|