38 results on '"Melvyn T. Chow"'
Search Results
2. Supplementary Figure 2 from Anti-PD-1 Antibody Therapy Potently Enhances the Eradication of Established Tumors By Gene-Modified T Cells
- Author
-
Phillip K. Darcy, Michael H. Kershaw, Mark J. Smyth, Melvyn T. Chow, Nicole M. Haynes, Paul A. Beavis, Carmen S. Yong, Connie P.M. Duong, Christel Devaud, and Liza B. John
- Abstract
Supplementary Figure 2 - PDF file 20K, Expression of PD-1 is upregulated on CAR T cells following antigen stimulation with e0771-Her2+ tumor cells
- Published
- 2023
- Full Text
- View/download PDF
3. upplementary Figure 3 from Anti-PD-1 Antibody Therapy Potently Enhances the Eradication of Established Tumors By Gene-Modified T Cells
- Author
-
Phillip K. Darcy, Michael H. Kershaw, Mark J. Smyth, Melvyn T. Chow, Nicole M. Haynes, Paul A. Beavis, Carmen S. Yong, Connie P.M. Duong, Christel Devaud, and Liza B. John
- Abstract
upplementary Figure 3 - PDF file 17K, Gr1+ cells express PD-1 and PD-L1
- Published
- 2023
- Full Text
- View/download PDF
4. Data from Anti-PD-1 Antibody Therapy Potently Enhances the Eradication of Established Tumors By Gene-Modified T Cells
- Author
-
Phillip K. Darcy, Michael H. Kershaw, Mark J. Smyth, Melvyn T. Chow, Nicole M. Haynes, Paul A. Beavis, Carmen S. Yong, Connie P.M. Duong, Christel Devaud, and Liza B. John
- Abstract
Purpose: To determine the antitumor efficacy and toxicity of a novel combination approach involving adoptive T-cell immunotherapy using chimeric antigen receptor (CAR) T cells with an immunomodulatory reagent for blocking immunosuppression.Experimental Design: We examined whether administration of a PD-1 blocking antibody could increase the therapeutic activity of CAR T cells against two different Her-2+ tumors. The use of a self-antigen mouse model enabled investigation into the efficacy, mechanism, and toxicity of this combination approach.Results: In this study, we first showed a significant increase in the level of PD-1 expressed on transduced anti-Her-2 CD8+ T cells following antigen-specific stimulation with PD-L1+ tumor cells and that markers of activation and proliferation were increased in anti-Her-2 T cells in the presence of anti-PD-1 antibody. In adoptive transfer studies in Her-2 transgenic recipient mice, we showed a significant improvement in growth inhibition of two different Her-2+ tumors treated with anti-Her-2 T cells in combination with anti-PD-1 antibody. The therapeutic effects observed correlated with increased function of anti-Her-2 T cells following PD-1 blockade. Strikingly, a significant decrease in the percentage of Gr1+ CD11b+ myeloid-derived suppressor cells (MDSC) was observed in the tumor microenvironment of mice treated with the combination therapy. Importantly, increased antitumor effects were not associated with any autoimmune pathology in normal tissue expressing Her-2 antigen.Conclusion: This study shows that specifically blocking PD-1 immunosuppression can potently enhance CAR T-cell therapy that has significant implications for potentially improving therapeutic outcomes of this approach in patients with cancer. Clin Cancer Res; 19(20); 5636–46. ©2013 AACR.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure Legends from Anti-PD-1 Antibody Therapy Potently Enhances the Eradication of Established Tumors By Gene-Modified T Cells
- Author
-
Phillip K. Darcy, Michael H. Kershaw, Mark J. Smyth, Melvyn T. Chow, Nicole M. Haynes, Paul A. Beavis, Carmen S. Yong, Connie P.M. Duong, Christel Devaud, and Liza B. John
- Abstract
Supplementary Figure Legends - PDF file 66K, Supplementary Figure legends
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 1 from Primary Tumor Hypoxia Recruits CD11b+/Ly6Cmed/Ly6G+ Immune Suppressor Cells and Compromises NK Cell Cytotoxicity in the Premetastatic Niche
- Author
-
Andreas Möller, Mark J. Smyth, David D. Bowtell, Belinda S. Parker, Erica K. Sloan, Daniel M. Andrews, Christina S.F. Wong, Heloise M. Halse, Anna Chen, Melvyn T. Chow, and Jaclyn Sceneay
- Abstract
PDF file - 296K, A)Regulation of Hif-1� and Hif-2alpha by hypoxia in breast cancer cells. B) Schematic of injection schedule
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 6 from NLRP3 Suppresses NK Cell–Mediated Responses to Carcinogen-Induced Tumors and Metastases
- Author
-
Mark J. Smyth, Andreas Möller, Jürg Tschopp, Helene Duret, Christina S.F. Wong, Christophe Paget, Jaclyn Sceneay, and Melvyn T. Chow
- Abstract
PDF file 3180K, Normal NK cell homeostasis in NLRP3-/- mice.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 5 from NLRP3 Suppresses NK Cell–Mediated Responses to Carcinogen-Induced Tumors and Metastases
- Author
-
Mark J. Smyth, Andreas Möller, Jürg Tschopp, Helene Duret, Christina S.F. Wong, Christophe Paget, Jaclyn Sceneay, and Melvyn T. Chow
- Abstract
PDF file 1235K, Caspase-1 and IL-1 do not affect the development of lung metastasis.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 2 from Primary Tumor Hypoxia Recruits CD11b+/Ly6Cmed/Ly6G+ Immune Suppressor Cells and Compromises NK Cell Cytotoxicity in the Premetastatic Niche
- Author
-
Andreas Möller, Mark J. Smyth, David D. Bowtell, Belinda S. Parker, Erica K. Sloan, Daniel M. Andrews, Christina S.F. Wong, Heloise M. Halse, Anna Chen, Melvyn T. Chow, and Jaclyn Sceneay
- Abstract
PDF file - 221K, A) Pre-metastatic niche induced by hypoxia increases melanoma metastasis. B) Factors identified in HCM of breast cancer cells
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure 1 from NLRP3 Suppresses NK Cell–Mediated Responses to Carcinogen-Induced Tumors and Metastases
- Author
-
Mark J. Smyth, Andreas Möller, Jürg Tschopp, Helene Duret, Christina S.F. Wong, Christophe Paget, Jaclyn Sceneay, and Melvyn T. Chow
- Abstract
PDF file-862K.NLRP3 inflammasome promotes the formation of MCA-induced fibrosarcomas in BALB/c mice.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 4 from Primary Tumor Hypoxia Recruits CD11b+/Ly6Cmed/Ly6G+ Immune Suppressor Cells and Compromises NK Cell Cytotoxicity in the Premetastatic Niche
- Author
-
Andreas Möller, Mark J. Smyth, David D. Bowtell, Belinda S. Parker, Erica K. Sloan, Daniel M. Andrews, Christina S.F. Wong, Heloise M. Halse, Anna Chen, Melvyn T. Chow, and Jaclyn Sceneay
- Abstract
PDF file - 49K, A) Analysis of other immune cell population in the pre-metastatic niche. B) CD69 expression on NK cells in pre-metastatic niche. C) Confirmation of NK cell ablation with anti-asialoGM1
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 3 from Primary Tumor Hypoxia Recruits CD11b+/Ly6Cmed/Ly6G+ Immune Suppressor Cells and Compromises NK Cell Cytotoxicity in the Premetastatic Niche
- Author
-
Andreas Möller, Mark J. Smyth, David D. Bowtell, Belinda S. Parker, Erica K. Sloan, Daniel M. Andrews, Christina S.F. Wong, Heloise M. Halse, Anna Chen, Melvyn T. Chow, and Jaclyn Sceneay
- Abstract
PDF file - 33K, A/B) Reduction of CD11b+/Ly6Cmed but not CD11b+/Ly6Chigh cells in pre-metastatic niche induced by HCM after MCP-1 neutralization
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figure 8 from NLRP3 Suppresses NK Cell–Mediated Responses to Carcinogen-Induced Tumors and Metastases
- Author
-
Mark J. Smyth, Andreas Möller, Jürg Tschopp, Helene Duret, Christina S.F. Wong, Christophe Paget, Jaclyn Sceneay, and Melvyn T. Chow
- Abstract
PDF file 1781 K, CD11b+ Gr-1int cells do not suppress or enhance NK cell cytotoxic function.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Methods and Materials and Figure Legends from NLRP3 Suppresses NK Cell–Mediated Responses to Carcinogen-Induced Tumors and Metastases
- Author
-
Mark J. Smyth, Andreas Möller, Jürg Tschopp, Helene Duret, Christina S.F. Wong, Christophe Paget, Jaclyn Sceneay, and Melvyn T. Chow
- Abstract
PDF file-166K
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure 9 from NLRP3 Suppresses NK Cell–Mediated Responses to Carcinogen-Induced Tumors and Metastases
- Author
-
Mark J. Smyth, Andreas Möller, Jürg Tschopp, Helene Duret, Christina S.F. Wong, Christophe Paget, Jaclyn Sceneay, and Melvyn T. Chow
- Abstract
PDF file 1.8 MB, Cytokine profile of CD11b+ Gr-1int cells.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Methods from Primary Tumor Hypoxia Recruits CD11b+/Ly6Cmed/Ly6G+ Immune Suppressor Cells and Compromises NK Cell Cytotoxicity in the Premetastatic Niche
- Author
-
Andreas Möller, Mark J. Smyth, David D. Bowtell, Belinda S. Parker, Erica K. Sloan, Daniel M. Andrews, Christina S.F. Wong, Heloise M. Halse, Anna Chen, Melvyn T. Chow, and Jaclyn Sceneay
- Abstract
PDF file - 90K
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figure Legends 1-4 from Primary Tumor Hypoxia Recruits CD11b+/Ly6Cmed/Ly6G+ Immune Suppressor Cells and Compromises NK Cell Cytotoxicity in the Premetastatic Niche
- Author
-
Andreas Möller, Mark J. Smyth, David D. Bowtell, Belinda S. Parker, Erica K. Sloan, Daniel M. Andrews, Christina S.F. Wong, Heloise M. Halse, Anna Chen, Melvyn T. Chow, and Jaclyn Sceneay
- Abstract
PDF file - 72K
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure 4 from NLRP3 Suppresses NK Cell–Mediated Responses to Carcinogen-Induced Tumors and Metastases
- Author
-
Mark J. Smyth, Andreas Möller, Jürg Tschopp, Helene Duret, Christina S.F. Wong, Christophe Paget, Jaclyn Sceneay, and Melvyn T. Chow
- Abstract
PDF file 806K, Host NLRP3 does not affect B16F10 subcutaneous growth.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figure 7 from NLRP3 Suppresses NK Cell–Mediated Responses to Carcinogen-Induced Tumors and Metastases
- Author
-
Mark J. Smyth, Andreas Möller, Jürg Tschopp, Helene Duret, Christina S.F. Wong, Christophe Paget, Jaclyn Sceneay, and Melvyn T. Chow
- Abstract
PDF file 2632K, CD11b+ Gr-1int cells express NLRP3.
- Published
- 2023
- Full Text
- View/download PDF
20. Chemokines and the immune response to cancer
- Author
-
Aleksandra J. Ozga, Andrew D. Luster, and Melvyn T. Chow
- Subjects
0301 basic medicine ,Chemokine ,Carcinogenesis ,animal diseases ,Immunology ,chemical and pharmacologic phenomena ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cell Movement ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Immunology and Allergy ,Malignant cells ,Tumor microenvironment ,Immunity ,Cancer ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Cancer treatment ,030104 developmental biology ,Infectious Diseases ,030220 oncology & carcinogenesis ,biology.protein ,bacteria ,Chemokines ,Immune cell migration ,Function (biology) - Abstract
Chemokines are chemotactic cytokines that regulate the migration of immune cells. Chemokines function as cues for the coordinated recruitment of immune cells into and out of tissue and also guide the spatial organization and cellular interactions of immune cells within tissues. Chemokines are critical in directing immune cell migration necessary to mount and then deliver an effective anti-tumor immune response; however, chemokines also participate in the generation and recruitment of immune cells that contribute to a pro-tumorigenic microenvironment. Here, we review the role of the chemokine system in anti-tumor and pro-tumor immune responses and discuss how malignant cells and the tumor microenvironment regulate the overall chemokine landscape to shape the type and outcome of immune responses to cancer and cancer treatment.
- Published
- 2021
- Full Text
- View/download PDF
21. CXCL10 chemokine regulates heterogeneity of the CD8+ T cell response and viral set point during chronic infection
- Author
-
Thorsten R. Mempel, Philippe Dehio, Andrew D. Luster, Aleksandra J. Ozga, Rachel L. Servis, Mateus E. Lopes, Mauro Di Pilato, Jeffrey Lian, and Melvyn T. Chow
- Subjects
White pulp ,Chemokine ,Receptors, CXCR3 ,Immunology ,CD8-Positive T-Lymphocytes ,Lymphocytic Choriomeningitis ,Biology ,Lymphocytic choriomeningitis ,CXCR3 ,Article ,B7-H1 Antigen ,Monocytes ,Mice ,medicine ,Animals ,Lymphocytic choriomeningitis virus ,Immunology and Allergy ,Cytotoxic T cell ,CXCL10 ,Hepatocyte Nuclear Factor 1-alpha ,Cell Self Renewal ,Clonal Selection, Antigen-Mediated ,Cell Proliferation ,Mice, Knockout ,Cell Differentiation ,medicine.disease ,Chemokine CXCL10 ,Mice, Inbred C57BL ,Chronic infection ,Infectious Diseases ,medicine.anatomical_structure ,Chronic Disease ,biology.protein ,Female ,Spleen ,CD8 - Abstract
CD8(+) T cells responding to chronic infection adapt an altered differentiation program that provides some restrain on pathogen replication yet limits immunopathology. This adaptation is imprinted in stem-like cells and propagated to their progeny. Understanding the molecular control of CD8(+) T cell differentiation in chronic infection has important therapeutic implications. Here, we found that the chemokine receptor CXCR3 was highly expressed on viral-specific stem-like CD8(+) T cells and that one of its ligands, CXCL10, regulated the persistence and heterogeneity of responding CD8(+) T cells in spleens of mice chronically infected with lymphocytic choriomeningitis virus. CXCL10 was produced by inflammatory monocytes and fibroblasts of the splenic red pulp where it granted stem-like cells access to signals promoting differentiation and limited their exposure to pro-survival niches in the white pulp. Consequently, functional CD8(+) T cell responses were greater in Cxcl10(−/−) mice and were associated with a lower viral set point.
- Published
- 2022
- Full Text
- View/download PDF
22. Type I NKT-cell-mediated TNF-α is a positive regulator of NLRP3 inflammasome priming
- Author
-
Daniel M. Andrews, Andreas Möller, Helene Duret, Melvyn T. Chow, Christophe Paget, Mark J. Smyth, and Christelle Faveeuw
- Subjects
Innate immune system ,integumentary system ,Immunology ,Priming (immunology) ,chemical and pharmacologic phenomena ,Inflammation ,Inflammasome ,Biology ,Natural killer T cell ,Cell biology ,AIM2 ,Immune system ,medicine ,Immunology and Allergy ,Tumor necrosis factor alpha ,medicine.symptom ,medicine.drug - Abstract
The NLRP3 inflammasome plays a crucial role in the innate immune response to pathogens and exogenous or endogenous danger signals. Its activity must be precisely and tightly regulated to generate tailored immune responses. However, the immune cell subsets and cytokines controlling NLRP3 inflammasome activity are still poorly understood. Here, we have shown a link between NKT-cell-mediated TNF-α and NLRP3 inflammasome activity. The NLRP3 inflammasome in APCs was critical to potentiate NKT-cell-mediated immune responses, since C57BL/6 NLRP3 inflammasome-deficient mice exhibited reduced responsiveness to α-galactosylceramide. Importantly, NKT cells were found to act as regulators of NLRP3 inflammasome signaling, as NKT-cell-derived TNF-α was required for optimal IL-1β and IL-18 production by myeloid cells in response to α-galactosylceramide, by acting on the NLRP3 inflammasome priming step. Thus, NKT cells play a role in the positive regulation of NLRP3 inflammasome priming by mediating the production of TNF-α, thus demonstrating another means by which NKT cells control early inflammation.
- Published
- 2014
- Full Text
- View/download PDF
23. The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions
- Author
-
Daniel M. Andrews, Christopher J. Chan, Marco Colonna, Melvyn T. Chow, Liam Town, Ludovic Martinet, Mark J. Smyth, David Ritchie, Susan Gilfillan, and Fernando Souza-Fonseca-Guimaraes
- Subjects
Antigens, Differentiation, T-Lymphocyte ,Cytotoxicity, Immunologic ,Lipopolysaccharides ,Lung Neoplasms ,CD96 ,CD226 ,medicine.medical_treatment ,Nectins ,Immunology ,Biology ,Natural killer cell ,Mice ,TIGIT ,Antigens, CD ,medicine ,Animals ,Immunology and Allergy ,CD155 ,Neoplasm Metastasis ,Receptors, Immunologic ,Receptor ,Cells, Cultured ,Mice, Knockout ,Cell adhesion molecule ,Neoplasms, Experimental ,Pneumonia ,Cell biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cytokine ,biology.protein ,Receptors, Virus ,Cell Adhesion Molecules ,Protein Binding - Abstract
CD96, CD226 (DNAM-1) and TIGIT belong to an emerging family of receptors that interact with nectin and nectin-like proteins. CD226 activates natural killer (NK) cell-mediated cytotoxicity, whereas TIGIT reportedly counterbalances CD226. In contrast, the role of CD96, which shares the ligand CD155 with CD226 and TIGIT, has remained unclear. In this study we found that CD96 competed with CD226 for CD155 binding and limited NK cell function by direct inhibition. As a result, Cd96(-/-) mice displayed hyperinflammatory responses to the bacterial product lipopolysaccharide (LPS) and resistance to carcinogenesis and experimental lung metastases. Our data provide the first description, to our knowledge, of the ability of CD96 to negatively control cytokine responses by NK cells. Blocking CD96 may have applications in pathologies in which NK cells are important.
- Published
- 2014
- Full Text
- View/download PDF
24. Intratumoral Activity of the CXCR3 Chemokine System Is Required for the Efficacy of Anti-PD-1 Therapy
- Author
-
Jennifer A. Lo, Melvyn T. Chow, Aleksandra J. Ozga, Genevieve M. Boland, Dennie T. Frederick, David E. Fisher, Andrew D. Luster, Gordon J. Freeman, and Rachel L. Servis
- Subjects
0301 basic medicine ,Chemokine ,Receptors, CXCR3 ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Immunology ,CD8-Positive T-Lymphocytes ,Biology ,Lymphocyte Activation ,CXCR3 ,Article ,Epigenesis, Genetic ,Immunomodulation ,Mice ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,stomatognathic system ,T-Lymphocyte Subsets ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Immunology and Allergy ,CXCL10 ,Molecular Targeted Therapy ,Mice, Knockout ,Tumor microenvironment ,Antibodies, Monoclonal ,Immunotherapy ,Xenograft Model Antitumor Assays ,Immune checkpoint ,Disease Models, Animal ,stomatognathic diseases ,030104 developmental biology ,Infectious Diseases ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,CXCL9 ,Chemokines ,Biomarkers ,CD8 - Abstract
Summary Despite compelling rates of durable clinical responses to programmed cell death-1 (PD-1) blockade, advances are needed to extend these benefits to resistant tumors. We found that tumor-bearing mice deficient in the chemokine receptor CXCR3 responded poorly to anti-PD-1 treatment. CXCR3 and its ligand CXCL9 were critical for a productive CD8+ T cell response in tumor-bearing mice treated with anti-PD-1 but were not required for the infiltration of CD8+ T cells into tumors. The anti-PD-1-induced anti-tumor response was facilitated by CXCL9 production from intratumoral CD103+ dendritic cells, suggesting that CXCR3 facilitates dendritic cell-T cell interactions within the tumor microenvironment. CXCR3 ligands in murine tumors and in plasma of melanoma patients were an indicator of clinical response to anti-PD-1, and their induction in non-responsive murine tumors promoted responsiveness to anti-PD-1. Our data suggest that the CXCR3 chemokine system is a biomarker for sensitivity to PD-1 blockade and that augmenting the intratumoral function of this chemokine system could improve clinical outcomes.
- Published
- 2019
- Full Text
- View/download PDF
25. Blockade of A 2A receptors potently suppresses the metastasis of CD73 + tumors
- Author
-
Liza B John, Christel Devaud, Paul A. Beavis, Christophe Paget, Mark J. Smyth, Melvyn T. Chow, Upulie Divisekera, Karen M. Dwyer, Phillip K. Darcy, and John Stagg
- Subjects
Cytotoxicity, Immunologic ,Receptor, Adenosine A2A ,medicine.medical_treatment ,T cell ,Biology ,Receptor, Adenosine A2B ,Granzymes ,Article ,Metastasis ,Mice ,Immune system ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Receptor ,5'-Nucleotidase ,Mice, Knockout ,Mice, Inbred BALB C ,Multidisciplinary ,Neoplasms, Experimental ,Immunotherapy ,Triazoles ,Flow Cytometry ,medicine.disease ,Adenosine receptor ,Adenosine A2 Receptor Antagonists ,Blockade ,Killer Cells, Natural ,Mice, Inbred C57BL ,Pyrimidines ,medicine.anatomical_structure ,Granzyme ,Xanthines ,Immunology ,Cancer research ,biology.protein - Abstract
CD73 inhibits antitumor immunity through the activation of adenosine receptors expressed on multiple immune subsets. CD73 also enhances tumor metastasis, although the nature of the immune subsets and adenosine receptor subtypes involved in this process are largely unknown. In this study, we revealed that A 2A /A 2B receptor antagonists were effective in reducing the metastasis of tumors expressing CD73 endogenously (4T1.2 breast tumors) and when CD73 was ectopically expressed (B16F10 melanoma). A 2A −/− mice were strongly protected against tumor metastasis, indicating that host A 2A receptors enhanced tumor metastasis. A 2A blockade enhanced natural killer (NK) cell maturation and cytotoxic function in vitro, reduced metastasis in a perforin-dependent manner, and enhanced NK cell expression of granzyme B in vivo, strongly suggesting that the antimetastatic effect of A 2A blockade was due to enhanced NK cell function. Interestingly, A 2B blockade had no effect on NK cell cytotoxicity, indicating that an NK cell-independent mechanism also contributed to the increased metastasis of CD73 + tumors. Our results thus revealed that CD73 promotes tumor metastasis through multiple mechanisms, including suppression of NK cell function. Furthermore, our data strongly suggest that A 2A or A 2B antagonists may be useful for the treatment of metastatic disease. Overall, our study has potential therapeutic implications given that A 2A /A 2B receptor antagonists have already entered clinical trials in other therapeutic settings.
- Published
- 2013
- Full Text
- View/download PDF
26. NLRP3 Suppresses NK Cell–Mediated Responses to Carcinogen-Induced Tumors and Metastases
- Author
-
Christina S.F. Wong, Helene Duret, Andreas Möller, Jaclyn Sceneay, Christophe Paget, Melvyn T. Chow, Mark J. Smyth, and Jürg Tschopp
- Subjects
Male ,Cancer Research ,Chemokine ,Adoptive cell transfer ,Lung Neoplasms ,Inflammasomes ,Fibrosarcoma ,Cell ,Population ,Melanoma, Experimental ,Mice, Transgenic ,Adenocarcinoma ,Biology ,medicine.disease_cause ,Chemokine CXCL9 ,CCL5 ,Mice ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Animals ,Myeloid Cells ,Neoplasm Metastasis ,education ,Chemokine CCL5 ,education.field_of_study ,CD11b Antigen ,integumentary system ,Melanoma ,Mammary Neoplasms, Experimental ,Prostatic Neoplasms ,Inflammasome ,Neoplasms, Experimental ,medicine.disease ,Killer Cells, Natural ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Oncology ,Immunology ,Cancer research ,biology.protein ,Carrier Proteins ,Carcinogenesis ,Methylcholanthrene ,medicine.drug - Abstract
The NLRP3 inflammasome acts as a danger signal sensor that triggers and coordinates the inflammatory response upon infectious insults or tissue injury and damage. However, the role of the NLRP3 inflammasome in natural killer (NK) cell–mediated control of tumor immunity is poorly understood. Here, we show in a model of chemical-induced carcinogenesis and a series of experimental and spontaneous metastases models that mice lacking NLRP3 display significantly reduced tumor burden than control wild-type (WT) mice. The suppression of spontaneous and experimental tumor metastases and methylcholanthrene (MCA)-induced sarcomas in mice deficient for NLRP3 was NK cell and IFN-γ–dependent. Focusing on the amenable B16F10 experimental lung metastases model, we determined that expression of NLRP3 in bone marrow–derived cells was necessary for optimal tumor metastasis. Tumor-driven expansion of CD11b+Gr-1intermediate (Gr-1int) myeloid cells within the lung tumor microenvironment of NLRP3−/− mice was coincident with increased lung infiltrating activated NK cells and an enhanced antimetastatic response. The CD11b+Gr-1int myeloid cells displayed a unique cell surface phenotype and were characterized by their elevated production of CCL5 and CXCL9 chemokines. Adoptive transfer of this population into WT mice enhanced NK cell numbers in, and suppression of, B16F10 lung metastases. Together, these data suggested that NLRP3 is an important suppressor of NK cell–mediated control of carcinogenesis and metastases and identify CD11b+Gr-1int myeloid cells that promote NK cell antimetastatic function. Cancer Res; 72(22); 5721–32. ©2012 AACR.
- Published
- 2012
- Full Text
- View/download PDF
27. Inflammation and immune surveillance in cancer
- Author
-
Melvyn T. Chow, Andreas Möller, and Mark J. Smyth
- Subjects
Cancer Research ,Inflammasomes ,Inflammation ,Biology ,Immune system ,Immunity ,Neoplasms ,Tumor Microenvironment ,medicine ,Humans ,Immunologic Factors ,HMGB1 Protein ,Immunologic Surveillance ,Toll-like receptor ,Tumor microenvironment ,Toll-Like Receptors ,Cancer ,Inflammasome ,medicine.disease ,Immunity, Innate ,Immunoediting ,Immunology ,Disease Progression ,Cytokines ,medicine.symptom ,medicine.drug - Abstract
Chronic inflammation is a risk factor for tumor development. However, understanding the effect of the immune system on tumor development has only been significantly advanced over the past two decades. We now appreciate that the immune system, in addition to tumor-suppressive function by eliminating nascent transformed tumor cells, can also exert selection pressure on tumor cells and facilitate tumor growth by providing a favorable tumor microenvironment. Yet, the distinctions between tumor-promoting inflammation and tumor-suppressive immunity are still not clear due to the dual role of some cytokines and other molecules in the immune system. The danger signal hypothesis has shaped our view of the role of immunity in cancer development, but still little is known about the exact role of danger signal receptors in cancer progression. In this review, we introduce the processes of cancer immunoediting and inflammation-induced cancer and discuss what is currently known about the role of danger signal receptors in cancer development and progression.
- Published
- 2012
- Full Text
- View/download PDF
28. Chemokines in cancer
- Author
-
Andrew D. Luster and Melvyn T. Chow
- Subjects
CCR1 ,Cancer Research ,Chemokine ,Tumor microenvironment ,Stromal cell ,biology ,Neovascularization, Pathologic ,Chemokine receptor CCR5 ,Immunology ,CCL18 ,Article ,Cell biology ,Neoplasms ,Cancer cell ,biology.protein ,Disease Progression ,Tumor Microenvironment ,Animals ,Humans ,CCR10 ,Immunotherapy ,Chemokines ,Neoplasm Metastasis - Abstract
Chemokines are chemotactic cytokines that control the migration of cells between tissues and the positioning and interactions of cells within tissue. The chemokine superfamily consists of approximately 50 endogenous chemokine ligands and 20 G protein–coupled seven-transmembrane spanning signaling receptors. Chemokines mediate the host response to cancer by directing the trafficking of leukocytes into the tumor microenvironment. This migratory response is complex and consists of diverse leukocyte subsets with both antitumor and protumor activities. Although chemokines were initially appreciated as important mediators of immune cell migration, we now know that they also play important roles in the biology of nonimmune cells important for tumor growth and progression. Chemokines can directly modulate the growth of tumors by inducing the proliferation of cancer cells and preventing their apoptosis. They also direct tumor cell movement required for metastasis. Chemokines can also indirectly modulate tumor growth through their effects on tumor stromal cells and by inducing the release of growth and angiogenic factors from cells in the tumor microenvironment. In this Masters of Immunology primer, we focus on recent advances in understanding the complex nature of the chemokine system in tumor biology with a focus on how the chemokine system could be used to augment cancer immunotherapeutic strategies to elicit a more robust and long-lasting host antitumor immune response. Cancer Immunol Res; 2(12); 1125–31. ©2014 AACR.
- Published
- 2014
29. Cancer cell–autonomous contribution of type I interferon signaling to the efficacy of chemotherapy
- Author
-
Aicha Goubar, Julien Adam, Joanna Cyrta, Kariman Chaba, Mireia Niso-Santano, Valentina La Sorsa, Laurence Zitvogel, Christina Pfirschke, Guido Kroemer, Gilles Uzé, Oliver Kepp, Magali Lacroix-Triki, Francesca Urbani, Antonella Sistigu, Elisa E. Baracco, Suzette Delaloge, Sylvain Ladoire, David Enot, Camilla Engblom, Marco Bianchi, Laurent Arnould, Virginie Quidville, Mauro Delorenzi, Enrico Proietti, Melvyn T. Chow, Laetitia Aymeric, Fabrice Andre, Giovanna Ziccheddu, Joachim L. Schultze, Alexander M.M. Eggermont, Thomas Tüting, Laetitia Fend, Mark J. Smyth, Catarina Remédios, Takahiro Yamazaki, Robert D. Schreiber, Laura Bracci, Erika Vacchelli, Rosa Conforti, Ilio Vitale, Paola Sestili, Mikael J. Pittet, Marie Charlotte Dessoliers, Filippo Belardelli, Vichnou Poirier-Colame, Frédérique Penault-Llorca, Jean Philippe Spano, Lajos Pusztai, Xavier Préville, Dalil Hannani, Yuting Ma, National Cancer Institute Regina Elena [Rome, Italy], Immunologie des tumeurs et immunothérapie (UMR 1015), Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), Apoptose, cancer et immunité (Equipe labellisée Ligue contre le cancer - CRC - Inserm U1138), Institut Gustave Roussy (IGR)-Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie - Paris 6 (UPMC)-École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC)-École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Immunologie et Cancérologie Intégratives (CRC - Inserm U1138), Centre de Recherche des Cordeliers (CRC), Institut Gustave Roussy (IGR), Biomarqueurs prédictifs et nouvelles stratégies moléculaires en thérapeutique anticancéreuse (U981), Département de biologie et pathologie médicales [Gustave Roussy], Apoptose, cancer et immunité (U848), Centre d'Investigation Clinique en Biotherapie des cancers (CIC 1428 , CBT 507 ), Institut Gustave Roussy (IGR)-Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Centro de Investigacion Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III [Madrid] (ISC), Institut Gustave Roussy (IGR)-Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Life and Medical Sciences Institute for Genomics and Immunoregulation, Universität Bonn = University of Bonn, Dept. of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità (ISS), CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Department of Breast Medical Oncology [Houston], The University of Texas M.D. Anderson Cancer Center [Houston], Pathologie mammaire, Département de médecine oncologique [Gustave Roussy], Institut Gustave Roussy (IGR)-Institut Gustave Roussy (IGR), Centre Jean Perrin [Clermont-Ferrand] (UNICANCER/CJP), UNICANCER, Centre Régional de Lutte contre le cancer Georges-François Leclerc [Dijon] (UNICANCER/CRLCC-CGFL), Département de Biologie et pathologie des tumeurs [Centre Georges-François Leclerc], UNICANCER-UNICANCER, Harvard Medical School [Boston] (HMS), Dynamique des interactions membranaires normales et pathologiques (DIMNP), Université Montpellier 1 (UM1)-Université Montpellier 2 - Sciences et Techniques (UM2)-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), Department of Cell Biology and Neurosciences, Université Paris-Sud - Paris 11 (UP11), Sistigu, A, Yamazaki, T, Vacchelli, E, Chaba, K, Enot, Dp, Adam, J, Vitale, I, Goubar, A, Baracco, Ee, Remédios, C, Fend, L, Hannani, D, Aymeric, L, Ma, Y, Niso Santano, M, Kepp, O, Schultze, Jl, Tüting, T, Belardelli, F, Bracci, L, La Sorsa, V, Ziccheddu, G, Sestili, P, Urbani, F, Delorenzi, M, Lacroix Triki, M, Quidville, V, Conforti, R, Spano, Jp, Pusztai, L, Poirier Colame, V, Delaloge, S, Penault Llorca, F, Ladoire, S, Arnould, L, Cyrta, J, Dessoliers, Mc, Eggermont, A, Bianchi, MARCO EMILIO, Pittet, M, Engblom, C, Pfirschke, C, Préville, X, Uzè, G, Schreiber, Rd, Chow, Mt, Smyth, Mj, Proietti, E, André, F, Kroemer, G, Zitvogel, L., Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Gustave Roussy (IGR)-Université Paris-Sud - Paris 11 (UP11), Université Pierre et Marie Curie - Paris 6 (UPMC)-École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC)-École pratique des hautes études (EPHE), Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-École pratique des hautes études (EPHE), University of Bonn, Istituto Superiore di Sanità, Service d'Oncologie médicale [CHU Pitié-Salpêtrière], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Istituto Superiore di Sanita', and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)
- Subjects
Myxovirus Resistance Proteins ,Messenger ,Receptor, Interferon alpha-beta ,Inbred C57BL ,chemotherapy ,Interferon alpha-beta ,Mice ,Interferon ,Receptors ,Anthracyclines ,Neoplasm Metastasis ,RIG-I ,Pattern recognition receptor ,Adaptor Proteins ,General Medicine ,Neoadjuvant Therapy ,3. Good health ,Gene Expression Regulation, Neoplastic ,Treatment Outcome ,Receptors, Pattern Recognition ,Interferon Type I ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Female ,Immunocompetence ,medicine.drug ,Receptor ,Signal Transduction ,Breast Neoplasms ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,Pattern Recognition ,Settore BIO/09 ,General Biochemistry, Genetics and Molecular Biology ,Paracrine signalling ,Immune system ,medicine ,CXCL10 ,Animals ,Humans ,cancer ,RNA, Messenger ,Autocrine signalling ,Neoplastic ,[SDV.IMM.IMM]Life Sciences [q-bio]/Immunology/Immunotherapy ,Toll-Like Receptor 3 ,Mice, Inbred C57BL ,Vesicular Transport ,Chemokine CXCL10 ,Adaptor Proteins, Vesicular Transport ,Gene Expression Regulation ,Doxorubicin ,Immunology ,TLR3 ,RNA - Abstract
International audience; The immune system is routinely confronted with cell death resulting from the physiological turnover of renewable tissues, as well as from pathological insults of several types. We hypothesize the existence of a mechanism that allows the immune system to discriminate between physiological and pathological instances of cell death, but the factors that determine whether cellular demise is perceived as a neutral, tolerogenic or immunogenic event remain unclear 1. Infectious insults are accompanied by so-called microbe-associated molecular patterns (MAMPs), i.e., viral or bacterial products that activate immune cells through a panel of pattern-recognition receptors (PRRs) 2. Moreover, intracellular pathogens generally trigger adaptive mechanisms aimed toward the re-establishment of homeosta-sis, including the unfolded protein response (UPR) and autophagy 3,4. In mammals, MAMPs coupled to the activation of stress responses Some of the anti-neoplastic effects of anthracyclines in mice originate from the induction of innate and T cell-mediated anticancer immune responses. Here we demonstrate that anthracyclines stimulate the rapid production of type I interferons (IFNs) by malignant cells after activation of the endosomal pattern recognition receptor Toll-like receptor 3 (TLR3). By binding to IFN- and IFN- receptors (IFNARs) on neoplastic cells, type I IFNs trigger autocrine and paracrine circuitries that result in the release of chemokine (C-X-C motif) ligand 10 (CXCL10). Tumors lacking Tlr3 or Ifnar failed to respond to chemotherapy unless type I IFN or Cxcl10, respectively, was artificially supplied. Moreover, a type I IFN-related signature predicted clinical responses to anthracycline-based chemotherapy in several independent cohorts of patients with breast carcinoma characterized by poor prognosis. Our data suggest that anthracycline-mediated immune responses mimic those induced by viral pathogens. We surmise that such 'viral mimicry' constitutes a hallmark of successful chemotherapy. npg
- Published
- 2014
- Full Text
- View/download PDF
30. CD3bright signals on γδ T cells identify IL-17A-producing Vγ6Vδ1+ T cells
- Author
-
François Trottein, Adam P Uldrich, Thierry Mallevaey, Christophe Paget, Paul J Neeson, Dale I. Godfrey, Geoffrey R. Hill, N. A. Gherardin, Paul A. Beavis, David Dombrowicz, Delphine Staumont-Sallé, Maya Hassane, Denis A. Mogilenko, N. K. Escalante, Mark J. Smyth, Fernando Souza-Fonseca-Guimaraes, Melvyn T. Chow, Helene Duret, David Ritchie, Gabrielle T. Belz, Derudas, Marie-Hélène, Centre d’Infection et d’Immunité de Lille - INSERM U 1019 - UMR 9017 - UMR 8204 (CIIL), Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille)-Centre National de la Recherche Scientifique (CNRS), Cancer immunology program, Peter MacCallum Cancer Center, Sir Peter MacCallum Department of Oncology and Department of Pathology, University of Melbourne, Department of Microbiology and Immunology, QIMR Berghofer Medical Research Institute, Récepteurs nucléaires, maladies cardiovasculaires et diabète - U 1011 (RNMCD), Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), European Genomic Institute of Diabetes (EGID), Service de dermatologie, Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Department of Immunology, University of Toronto, Department of Bone Marrow Transplantation, Royal Brisbane Hospital, Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research (WEHI), School of Medicine, University of Queensland [Brisbane], his work was supported by an NHMRC Program Grant (454569 and 1013367). CP was supported by a postdoctoral fellowship from the US Department of Defense (W81XWH‐11‐1‐0585) and INSERM. MJS and FSFG were supported by a National Health and Medical Research Council of Australia (NHMRC) Australia Fellowship and Program Grant (1013367). MTC was supported by a Cancer Research Institute PhD scholarship. GTB was supported by an ARC Future Fellowship. FT was supported by CNRS. PN was supported by a National Health and Medical Research Council of Australia (NHMRC) Program Grant (1013367). DIG was supported by an NHMRC Senior Principal Research Fellowship (1020770). NAG was supported by a Leukaemia Foundation of Australia postgraduate scholarship. TM is supported by research grants from the Canadian Institutes of Health Research and the Crohn and Colitis Foundation of Canada, as well as a Tier 2 Canada Research Chair. NKE is a recipient of a Vanier Canada Graduate Scholarship. DAM, DS‐S and DD were supported by the Agence Nationale de la Recherche (ANR‐10‐LABX‐46) and the ‘Fondation de France’., We thank Josette Fontaine, Shin‐Foong Ngiow, Deborah Knight, Sébastien Fleury, Julien Wartelle and Kim Steegh for technical assistance. We thank Qerime Mundrea, Ben Venville, Jessica May, Joanne Sutton and Liam Town for maintaining and caring for the mice. We also thank the Peter MacCallum flow cytometry core facility for technical assistance., Institut Européen de Génomique du Diabète - European Genomic Institute for Diabetes - FR 3508 (EGID), Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre National de la Recherche Scientifique (CNRS), Centre National de la Recherche Scientifique (CNRS)-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille)-Université de Lille-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP), and Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
Male ,CD3 Complex ,Inflammasomes ,T-Lymphocytes ,Interleukin-1beta ,MESH: Amino Acid Sequence ,MESH: Interleukin-1beta/metabolism ,Interleukin-23 ,MESH: Lung/drug effects ,MESH: Skin/immunology ,MESH: Lung/immunology ,MESH: CD3 Complex/chemistry ,MESH: Lymphocyte Subsets/immunology ,Immunology and Allergy ,Homeostasis ,MESH: Animals ,IL-2 receptor ,MESH: Homeostasis/drug effects ,Lung ,MESH: Immunity ,Skin ,MESH: Interleukin-23 ,Orphan receptor ,education.field_of_study ,Imiquimod ,biology ,MESH: Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism ,Interleukin-17 ,Receptors, Antigen, T-Cell, gamma-delta ,MESH: T-Lymphocytes/immunology ,Nuclear Receptor Subfamily 1, Group F, Member 3 ,MESH: CD3 Complex/metabolism ,3. Good health ,MESH: NLR Family, Pyrin Domain-Containing 3 Protein ,medicine.anatomical_structure ,Phenotype ,MESH: Aminoquinolines/pharmacology ,MESH: Carrier Proteins/metabolism ,Aminoquinolines ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,MESH: Interleukin-17/biosynthesis ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,MESH: Skin/drug effects ,CD3 ,T cell ,MESH: T-Lymphocytes/drug effects ,Immunology ,Population ,Molecular Sequence Data ,MESH: Germ Cells/drug effects ,MESH: Imiquimod ,MESH: Phenotype ,Immune system ,Antigen ,MESH: Mice, Inbred C57BL ,NLR Family, Pyrin Domain-Containing 3 Protein ,MESH: Inflammasomes/drug effects ,medicine ,Animals ,Amino Acid Sequence ,education ,MESH: Molecular Sequence Data ,Innate immune system ,MESH: Inflammasomes/metabolism ,Immunity ,Cell Biology ,MESH: Lymphocyte Subsets/drug effects ,MESH: Male ,Lymphocyte Subsets ,Mice, Inbred C57BL ,Germ Cells ,MESH: Receptors, Antigen, T-Cell, gamma-delta/metabolism ,biology.protein ,Carrier Proteins - Abstract
International audience; Interleukin-17A (IL-17A) is a pro-inflammatory cytokine that has an important role at mucosal sites in a wide range of immune responses including infection, allergy and auto-immunity. γδ T cells are recognized as IL-17 producers, but based on the level of CD3 expression, we now define the remarkable ability of a CD3(bright) γδ T-cell subset with an effector memory phenotype to rapidly produce IL-17A, but not interferon-γ. CD3(bright) γδ T cells uniformly express the canonical germline encoded Vγ6/Vδ1(+) T-cell receptor. They are widely distributed with a preferential representation in the lungs and skin are negatively impacted in the absence of retinoic acid receptor-related orphan receptor gammat expression or endogenous flora. This population responded rapidly to various stimuli in a mechanism involving IL-23 and NOD-like receptor family, pyrin domain containing 3 (NLRP3)-inflammasome-dependent IL-1β. Finally, we demonstrated that IL-17-producing CD3(bright) γδ T cells responded promptly and strongly to pneumococcal infection and during skin inflammation. Here, we propose a new way to specifically analyze IL-17-producing Vγ6/Vδ1(+) T cells based on the level of CD3 signals. Using this gating strategy, our data reinforce the crucial role of this γδ T-cell subset in respiratory and skin disorders.
- Published
- 2014
- Full Text
- View/download PDF
31. Type I NKT-cell-mediated TNF-α is a positive regulator of NLRP3 inflammasome priming
- Author
-
Melvyn T, Chow, Helene, Duret, Daniel M, Andrews, Christelle, Faveeuw, Andreas, Möller, Mark J, Smyth, and Christophe, Paget
- Subjects
Inflammation ,Male ,Mice, Inbred C57BL ,Mice ,Tumor Necrosis Factor-alpha ,NLR Family, Pyrin Domain-Containing 3 Protein ,Animals ,Antigen-Presenting Cells ,Cytokines ,Natural Killer T-Cells ,Galactosylceramides ,Carrier Proteins - Abstract
The NLRP3 inflammasome plays a crucial role in the innate immune response to pathogens and exogenous or endogenous danger signals. Its activity must be precisely and tightly regulated to generate tailored immune responses. However, the immune cell subsets and cytokines controlling NLRP3 inflammasome activity are still poorly understood. Here, we have shown a link between NKT-cell-mediated TNF-α and NLRP3 inflammasome activity. The NLRP3 inflammasome in APCs was critical to potentiate NKT-cell-mediated immune responses, since C57BL/6 NLRP3 inflammasome-deficient mice exhibited reduced responsiveness to α-galactosylceramide. Importantly, NKT cells were found to act as regulators of NLRP3 inflammasome signaling, as NKT-cell-derived TNF-α was required for optimal IL-1β and IL-18 production by myeloid cells in response to α-galactosylceramide, by acting on the NLRP3 inflammasome priming step. Thus, NKT cells play a role in the positive regulation of NLRP3 inflammasome priming by mediating the production of TNF-α, thus demonstrating another means by which NKT cells control early inflammation.
- Published
- 2013
32. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells
- Author
-
Carmen S M Yong, Melvyn T. Chow, Liza B John, Connie P.M. Duong, Phillip K. Darcy, Christel Devaud, Paul A. Beavis, Mark J. Smyth, Michael H. Kershaw, and Nicole M. Haynes
- Subjects
Cancer Research ,Adoptive cell transfer ,Receptor, ErbB-2 ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Antineoplastic Agents ,Autoimmunity ,Mice, Transgenic ,Lymphocyte Activation ,chemistry.chemical_compound ,Mice ,Antigen ,T-Lymphocyte Subsets ,Cell Line, Tumor ,Neoplasms ,Blocking antibody ,Medicine ,Animals ,Humans ,Myeloid Cells ,Antigens ,Tumor microenvironment ,business.industry ,Antibodies, Monoclonal ,Immunotherapy ,Adoptive Transfer ,Chimeric antigen receptor ,Tumor Burden ,Disease Models, Animal ,Oncology ,chemistry ,Immunology ,Cancer research ,Growth inhibition ,business ,CD8 - Abstract
Purpose: To determine the antitumor efficacy and toxicity of a novel combination approach involving adoptive T-cell immunotherapy using chimeric antigen receptor (CAR) T cells with an immunomodulatory reagent for blocking immunosuppression. Experimental Design: We examined whether administration of a PD-1 blocking antibody could increase the therapeutic activity of CAR T cells against two different Her-2+ tumors. The use of a self-antigen mouse model enabled investigation into the efficacy, mechanism, and toxicity of this combination approach. Results: In this study, we first showed a significant increase in the level of PD-1 expressed on transduced anti-Her-2 CD8+ T cells following antigen-specific stimulation with PD-L1+ tumor cells and that markers of activation and proliferation were increased in anti-Her-2 T cells in the presence of anti-PD-1 antibody. In adoptive transfer studies in Her-2 transgenic recipient mice, we showed a significant improvement in growth inhibition of two different Her-2+ tumors treated with anti-Her-2 T cells in combination with anti-PD-1 antibody. The therapeutic effects observed correlated with increased function of anti-Her-2 T cells following PD-1 blockade. Strikingly, a significant decrease in the percentage of Gr1+ CD11b+ myeloid-derived suppressor cells (MDSC) was observed in the tumor microenvironment of mice treated with the combination therapy. Importantly, increased antitumor effects were not associated with any autoimmune pathology in normal tissue expressing Her-2 antigen. Conclusion: This study shows that specifically blocking PD-1 immunosuppression can potently enhance CAR T-cell therapy that has significant implications for potentially improving therapeutic outcomes of this approach in patients with cancer. Clin Cancer Res; 19(20); 5636–46. ©2013 AACR . See related article by Morales-Kastresana, et al., [p. 5546][1] This article is featured in Highlights of This Issue, [p. 5543][2] [1]: /lookup/volpage/19/5546?iss=20 [2]: /lookup/volpage/19/5543?iss=20
- Published
- 2013
33. NLRP3 promotes inflammation-induced skin cancer but is dispensable for asbestos-induced mesothelioma
- Author
-
Jürg Tschopp, Mark J. Smyth, Andreas Möller, and Melvyn T. Chow
- Subjects
Mesothelioma ,Skin Neoplasms ,medicine.medical_treatment ,Pleural Neoplasms ,Immunology ,Interleukin-1beta ,Inflammation ,medicine.disease_cause ,Asbestos ,03 medical and health sciences ,Peritoneal cavity ,Mice ,0302 clinical medicine ,Cell Movement ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Immunology and Allergy ,Animals ,Humans ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Mice, Inbred BALB C ,integumentary system ,Papilloma ,business.industry ,Cancer ,Inflammasome ,Cell Biology ,Environmental Exposure ,medicine.disease ,3. Good health ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,Cytokine ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,Female ,Skin cancer ,medicine.symptom ,Peritoneum ,business ,Carrier Proteins ,medicine.drug - Abstract
Asbestos exposure can result in serious and frequently lethal diseases including malignant mesothelioma. The host sensor for asbestos induced inflammation is the NLRP3 inflammasome and it is widely assumed that this complex is essential for asbestos induced cancers. Here we report that acute interleukin 1beta production and recruitment of immune cells into peritoneal cavity were significantly decreased in the NLRP3 deficient mice after the administration of asbestos. However NLRP3 deficient mice displayed a similar incidence of malignant mesothelioma and survival times as wild type mice. Thus early inflammatory reactions triggered by asbestos are NLRP3 dependent but NLRP3 is not critical in the chronic development of asbestos induced mesothelioma. Notably in a two stage carcinogenesis induced papilloma model NLRP3 deficient mice showed a resistance phenotype in two different strain backgrounds suggesting a tumour promoting role of NLRP3 in certain chemically induced cancer types.Immunology and Cell Biology advance online publication 25 September 2012; doi:10.1038/icb.2012.46.
- Published
- 2012
34. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche
- Author
-
Anna Chen, Erica K. Sloan, Andreas Möller, Christina S.F. Wong, Daniel M. Andrews, Mark J. Smyth, Melvyn T. Chow, Belinda S. Parker, David D.L. Bowtell, Jaclyn Sceneay, and Heloise Halse
- Subjects
Cytotoxicity, Immunologic ,Cancer Research ,Myeloid ,Lung Neoplasms ,Angiogenesis ,Melanoma, Experimental ,Mice, Transgenic ,Biology ,Mice ,Immune system ,Cell Line, Tumor ,medicine ,Cytotoxic T cell ,Animals ,Antigens, Ly ,Mammary tumor ,CD11b Antigen ,Melanoma ,Mammary Neoplasms, Experimental ,medicine.disease ,Primary tumor ,Cell Hypoxia ,Killer Cells, Natural ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Oncology ,Immunology ,Cancer research ,Female ,Bone marrow ,Precancerous Conditions - Abstract
Hypoxia within a tumor acts as a strong selective pressure that promotes angiogenesis, invasion, and metastatic spread. In this study, we used immune competent bone marrow chimeric mice and syngeneic orthotopic mammary cancer models to show that hypoxia in the primary tumor promotes premetastatic niche formation in secondary organs. Injection of mice with cell-free conditioned medium derived from hypoxic mammary tumor cells resulted in increased bone marrow–derived cell infiltration into the lung in the absence of a primary tumor and led to increased metastatic burden in mammary and melanoma experimental metastasis models. By characterizing the composition of infiltrating bone marrow–derived cells, we identified CD11b+/Ly6Cmed/Ly6G+ myeloid and CD3−/NK1.1+ immune cell lineages as key constituents of the premetastatic niche. Furthermore, the cytotoxicity of natural killer (NK) cells was significantly decreased, resulting in a reduced antitumor response that allowed metastasis formation in secondary organs to a similar extent as ablation of NK cells. In contrast, metastatic burden was decreased when active NK cells were present in premetastatic lungs. Together, our findings suggest that primary tumor hypoxia provides cytokines and growth factors capable of creating a premetastatic niche through recruitment of CD11b+/Ly6Cmed/Ly6G+ myeloid cells and a reduction in the cytotoxic effector functions of NK cell populations. Cancer Res; 72(16); 3906–11. ©2012 AACR.
- Published
- 2012
35. Role of γδ T cells in α-galactosylceramide-mediated immunity
- Author
-
Christophe Paget, Helene Duret, Mark J. Smyth, Melvyn T. Chow, and Stephen R. Mattarollo
- Subjects
Cytotoxicity, Immunologic ,Lung Neoplasms ,Lymphoma, B-Cell ,T cell ,T-Lymphocytes ,Immunology ,Melanoma, Experimental ,chemical and pharmacologic phenomena ,Galactosylceramides ,Biology ,Adaptive Immunity ,Lymphocyte Activation ,Interleukin 21 ,Carcinoma, Lewis Lung ,Mice ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,IL-2 receptor ,Antigen-presenting cell ,Mice, Knockout ,Interleukin-18 ,Receptors, Antigen, T-Cell, gamma-delta ,Dendritic cell ,Natural killer T cell ,Interleukin-12 ,Immunity, Innate ,carbohydrates (lipids) ,Killer Cells, Natural ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Liver ,Interleukin 12 ,lipids (amino acids, peptides, and proteins) ,Antigens, CD1d ,Spleen ,Protein Binding - Abstract
Attempts to harness mouse type I NKT cells in different therapeutic settings including cancer, infection, and autoimmunity have proven fruitful using the CD1d-binding glycolipid α-galactosylceramide (α-GalCer). In these different models, the effects of α-GalCer mainly relied on the establishment of a type I NKT cell-dependent immune cascade involving dendritic cell, NK cell, B cell, or conventional CD4+ and CD8+ T cell activation/regulation as well as immunomodulatory cytokine production. In this study, we showed that γδ T cells, another population of innate-like T lymphocytes, displayed a phenotype of activated cells (cytokine production and cytotoxic properties) and were required to achieve an optimal α-GalCer–induced immune response. Using gene-targeted mice and recombinant cytokines, a critical need for IL-12 and IL-18 has been shown in the α-GalCer–induced IFN-γ production by γδ T cells. Moreover, this cytokine production occurred downstream of type I NKT cell response, suggesting their bystander effect on γδ T cells. In line with this, γδ T cells failed to directly recognize the CD1d/α-GalCer complex. We also provided evidence that γδ T cells increase their cytotoxic properties after α-GalCer injection, resulting in an increase in killing of tumor cell targets. Moreover, using cancer models, we demonstrated that γδ T cells were required for an optimal α-GalCer–mediated anti-tumor activity. Finally, we reported that immunization of wild-type mice with α-GalCer enhanced the adaptive immune response elicited by OVA, and this effect was strongly mediated by γδ T cells. We conclude that γδ T cells amplify the innate and acquired response to α-GalCer, with possibly important outcomes for the therapeutic effects of this compound.
- Published
- 2012
36. Homeostatic defects in interleukin 18-deficient mice contribute to protection against the lethal effects of endotoxin
- Author
-
Joseph A. Trapani, Claire L Cotterell, Shizuo Akira, Yoichiro Iwakura, Yuting Ma, Mark J. Smyth, Desiree A Anthony, Laurence Zitvogel, Daniel M. Andrews, Sally V. Watt, and Melvyn T. Chow
- Subjects
Lipopolysaccharides ,T cell ,T-Lymphocytes ,Immunology ,Biology ,Interleukin 21 ,Interferon-gamma ,Mice ,medicine ,Immunology and Allergy ,Animals ,Homeostasis ,Cells, Cultured ,Inflammation ,Mice, Knockout ,Innate immune system ,Lymphokine-activated killer cell ,Innate lymphoid cell ,Interleukin-17 ,Interleukin-18 ,Interleukin ,Receptors, Antigen, T-Cell, gamma-delta ,Cell Biology ,Cell biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Interleukin 15 ,Interleukin 12 - Abstract
Toll-like receptor-4-lipopolysaccharide (LPS)-mediated inflammation is used to delineate signals involved in cross-talk between antigen-presenting cells (APCs) and lymphocytes such as natural killer (NK) cells. Following APC stimulation and cytokine release, NK cells produce interferon (IFN)-γ. High levels of LPS induce endotoxicosis, a systemic inflammatory disease in which IFN-γ causes significant morbidity and mortality. Several studies have highlighted the role of interleukin (IL)-18, IL-1β, IL-17A and IFN-γ in the development of endotoxicosis, but whether these cytokines interact with each other is yet to be determined. Our data demonstrate that IL-18 and IL-17A have important roles in NK cell IFN-γ production during endotoxicosis. Importantly, we provide the first evidence that IL-18 also has a role in IL-17A production by T-cell receptor (TCR)-δ cells. Furthermore, we demonstrate that IL-18-deficient mice have a defect in γδ T-cell homeostasis and IL-1β production, both of which can contribute to the development of disease through induction of IL-17A. These results reveal novel requirements for IL-18 in innate immune cell homeostasis and activation, demonstrating that the role of IL-18 in innate immunity occurs at a level other than activation.
- Published
- 2011
37. A role for granzyme M in TLR4-driven inflammation and endotoxicosis
- Author
-
Phillip I. Bird, Shizou Akira, Sally V. Watt, Desiree A Anthony, Melvyn T. Chow, Joseph A. Trapani, Mark J. Smyth, Daniel M. Andrews, and Colin M. House
- Subjects
Lipopolysaccharides ,medicine.medical_treatment ,Immunology ,Inflammation ,Biology ,Ligands ,Granzymes ,Mice ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Myeloid Cells ,Mice, Knockout ,Shock, Septic ,Immunity, Innate ,Cell biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,Toll-Like Receptor 4 ,Cytokine ,Granzyme ,Perforin ,TLR4 ,biology.protein ,Tumor necrosis factor alpha ,medicine.symptom ,Granzyme M ,Inflammation Mediators ,Signal Transduction - Abstract
Lymphocyte perforin and serine protease granzymes are well-recognized extrinsic mediators of apoptosis. We now demonstrate that cytotoxic lymphocyte granule components profoundly augment the myeloid cell inflammatory cytokine cascade in response to TLR4 ligation. Whereas caspase-1–deficient mice were completely resistant to LPS, reduced serum cytokine production and resistance to lethal endotoxicosis were also obtained with perforin-deficient mice, indicating a role for granzymes. Consistently, a lack of granzyme M (GrzM) resulted in reduced serum IL-1α, IL-1β, TNF, and IFN-γ levels and significantly reduced susceptibility to lethal endotoxicosis. These altered responses were also observed in granzyme A-deficient but not granzyme B-deficient mice. A role for APC–NK cell cross-talk in the inflammatory cascade was highlighted, as GrzM was exclusively expressed by NK cells and resistance to LPS was also observed on a RAG-1/GrzM-double deficient background. Collectively, the data suggest that NK cell GrzM augments the inflammatory cascade downstream of LPS-TLR4 signaling, which ultimately results in lethal endotoxicosis. Most importantly, these data demonstrate that granzymes should no longer be considered solely as mediators of apoptosis, but additionally as potential key regulators of inflammation.
- Published
- 2010
38. Toll-like receptor 3 regulates NK cell responses to cytokines and controls experimental metastasis
- Author
-
Camille Guillerey, Stuart D. Olver, Kim Miles, Andreas Möller, Melvyn T. Chow, Kazuyoshi Takeda, Mark J. Smyth, and Jaclyn Sceneay
- Subjects
Toll-like receptor ,Lymphokine-activated killer cell ,viruses ,medicine.medical_treatment ,Janus kinase 3 ,Immunology ,virus diseases ,hemic and immune systems ,chemical and pharmacologic phenomena ,Biology ,Interleukin 21 ,Cytokine ,Immune system ,Oncology ,TLR3 ,Interleukin 12 ,Cancer research ,medicine ,Immunology and Allergy ,Original Research - Abstract
The Toll-like receptor 3 (TLR3) agonist poly(I:C) is a promising adjuvant for cancer vaccines due to its induction of potent antitumor responses occurring primarily through the activation of dendritic cells (DCs) and natural killer (NK) cells. However, little is known about the role of TLR3 sensing of endogenous ligands in innate tumor immunosurveillance. Here, we investigated whether TLR3 could modulate immune responses and facilitate tumor control without administration of an agonist. We observed only limited impact of TLR3 deficiency on spontaneous carcinogenesis and primary growth of B16F10, E0771 or MC38 tumors when injected subcutaneously to mice. Nevertheless, TLR3 was observed to limit experimental B16F10 lung metastasis, an immunologic constraint dependent on both IFNγ secretion and NK cells. Interestingly, we observed that NK cells derived from Tlr3 null (Tlr3-/- ) mice were hyporesponsive to cytokine stimulation. Indeed, compared with NK cells with intact TLR3, Tlr3-/- NK cells produced significantly reduced pro-inflammatory cytokines, including IFNγ, when incubated in the presence of different combinations of IL-12, IL-18 and IL-15. Bone-marrow chimera experiments established that competent NK cell responses required TLR3 sensing on radio-sensitive immune cells. Intriguingly, although CD8α DCs robustly express high levels of TLR3, we found that those cells were not necessary for efficient IFNγ production by NK cells. Moreover, the defective NK cell phenotype of Tlr3-/- mice appeared to be independent of the gut microbiota. Altogether, our data demonstrate a pivotal role of endogenous TLR3 stimulation for the acquisition of full NK cell functions and immune protection against experimental metastasis.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.