1
|
Liu F, Wang Y, Yu J. Role of inflammation and immune response in atherosclerosis: Mechanisms, modulations, and therapeutic targets. Hum Immunol 2023; 84:439-449. [PMID: 37353446 DOI: 10.1016/j.humimm.2023.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/19/2023] [Accepted: 06/08/2023] [Indexed: 06/25/2023]
Abstract
Cardiovascular diseases (CVDs) have emerged as the leading cause of mortality globally, with atherosclerosis being a prominent focus of investigation among medical researchers worldwide. Atherosclerosis is characterized as a disease of the large and medium-sized arteries that is multifocal, accumulative, and immunoinflammatory in nature, resulting from the deposition of lipids. Accumulating evidence suggests that inflammatory responses and immunoregulation play a vital role in the occurrence and development of atherosclerosis. While existing treatments for atherosclerosis can assist in symptom management and slowing disease progression, a complete cure remains elusive. Consequently, there is significant interest in research and development of potential new drugs for this condition. Therefore, this review aims to consolidate the current understanding of the pathogenesis of atherosclerosis with an emphasis on inflammation, immune response and infection. Besides, it examines the effects and mechanisms of immunological modulations in atherosclerosis, and the potential therapeutic targets and drugs for intervening in the inflammatory responses and immunoregulation associated with atherosclerosis. Additionally, novel drug options for treating atherosclerosis are explored within the context of this review.
Collapse
Affiliation(s)
- Fang Liu
- Department of Vascular Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; International Genome Center, Jiangsu University, Zhenjiang 212013, China.
| | - Yijun Wang
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| | - Jiayin Yu
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
2
|
Ji Y, Madrasi K, Knee DA, Gruenbaum L, Apgar JF, Burke JM, Gomes B. Quantitative systems pharmacology model of GITR-mediated T cell dynamics in tumor microenvironment. CPT Pharmacometrics Syst Pharmacol 2023; 12:413-424. [PMID: 36710369 PMCID: PMC10014051 DOI: 10.1002/psp4.12925] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 01/31/2023] Open
Abstract
T cell interaction in the tumor microenvironment is a key component of immuno-oncology therapy. Glucocorticoid-induced tumor necrosis factor receptor (TNFR)-related protein (GITR) is expressed on immune cells including regulatory T cells (Tregs) and effector T cells (Teffs). Preclinical data suggest that agonism of GITR in combination with Fc-γ receptor-mediated depletion of Tregs results in increased intratumoral Teff:Treg ratio and tumor shrinkage. A novel quantitative systems pharmacology (QSP) model was developed for the murine anti-GITR agonist antibody, DTA-1.mIgG2a, to describe the kinetics of intratumoral Tregs and Teffs in Colon26 and A20 syngeneic mouse tumor models. It adequately captured the time profiles of intratumoral Treg and Teff and serum DTA-1.mIgG2a and soluble GITR concentrations in both mouse models, and described the response differences between the two models. The QSP model provides a quantitative understanding of the trade-off between maximizing Treg depletion versus Teff agonism, and offers insights to optimize drug design and dose regimen.
Collapse
Affiliation(s)
- Yan Ji
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | - Deborah A Knee
- Novartis Institutes for Biomedical Research, San Diego, California, USA
| | - Lore Gruenbaum
- Therapy Acceleration Program, The Leukemia & Lymphoma Society, Rye Brook, New York, USA
| | | | - John M Burke
- Applied Biomath LLC, Concord, Massachusetts, USA
| | - Bruce Gomes
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| |
Collapse
|
3
|
Ji Y, Knee D, Chen X, Dang A, Mataraza J, Wolf B, Sy SKB. Model-informed drug development for immuno-oncology agonistic anti-GITR antibody GWN323: Dose selection based on MABEL and biologically active dose. Clin Transl Sci 2022; 15:2218-2229. [PMID: 35731955 PMCID: PMC9468570 DOI: 10.1111/cts.13355] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 01/25/2023] Open
Abstract
GWN323, an agonistic human anti-GITR (glucocorticoid-induced TNFR-related protein) IgG1 antibody, was studied clinically as an immuno-oncology therapeutic agent. A model-based minimum anticipated biological effect level (MABEL) approach integrating in vitro and in vivo data informed dose selection for the first-in-human (FIH) study. Data evaluated included pharmacokinetics (PK) of DTA-1.mIgG2a (mouse surrogate GITR antibody for GWN323), target-engagement pharmacodynamic (PD) marker soluble GITR (sGITR), tumor shrinkage in Colon26 syngeneic mice administered with DTA-1.mIgG2a, cytokine release of GWN323 in human peripheral blood mononuclear cells, and GITR binding affinity. A PK model was developed to describe DTA-1.mIgG2a PK, and its relationship with sGITR was also modeled. Human GWN323 PK was predicted by allometric scaling of mouse PK. Based on the totality of PK/PD modeling and in vitro and in vivo pharmacology and toxicology data, MABEL was estimated to be 3-10 mg once every 3 weeks (Q3W), which informed the starting dose selection of the FIH study. Based on tumor kinetic PK/PD modeling of tumor inhibition by DTA-1.mIgG2a in Colon26 mice and the predicted human PK of GWN323, the biologically active dose of GWN323 was predicted to be 350 mg Q3W, which informed the dose escalation of the FIH study. GWN323 PK from the FIH study was described by a population PK model; the relationship with ex vivo interleukin-2 release, a target-engagement marker, was also modeled. The clinical PK/PD modeling data supported the biological active dose projected from the translational PK/PD modeling in a "learn and confirm" paradigm of model-informed drug development of GWN323.
Collapse
Affiliation(s)
- Yan Ji
- Novartis Pharmaceuticals CorporationEast HanoverNew JerseyUSA
| | - Deborah Knee
- Novartis Institutes for BioMedical ResearchSan DiegoCaliforniaUSA
| | - Xinhui Chen
- Novartis Pharmaceuticals CorporationEast HanoverNew JerseyUSA
| | - Anhthu Dang
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
| | - Jennifer Mataraza
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
| | - Babette Wolf
- Novartis Institutes for BioMedical ResearchBaselSwitzerland
| | | |
Collapse
|
4
|
Painter JD, Akbari O. Type 2 Innate Lymphoid Cells: Protectors in Type 2 Diabetes. Front Immunol 2021; 12:727008. [PMID: 34489979 PMCID: PMC8416625 DOI: 10.3389/fimmu.2021.727008] [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: 06/17/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022] Open
Abstract
Type 2 innate lymphoid cells (ILC2) are the innate counterparts of Th2 cells and are critically involved in the maintenance of homeostasis in a variety of tissues. Instead of expressing specific antigen receptors, ILC2s respond to external stimuli such as alarmins released from damage. These cells help control the delicate balance of inflammation in adipose tissue, which is a determinant of metabolic outcome. ILC2s play a key role in the pathogenesis of type 2 diabetes mellitus (T2DM) through their protective effects on tissue homeostasis. A variety of crosstalk takes place between resident adipose cells and ILC2s, with each interaction playing a key role in controlling this balance. ILC2 effector function is associated with increased browning of adipose tissue and an anti-inflammatory immune profile. Trafficking and maintenance of ILC2 populations are critical for tissue homeostasis. The metabolic environment and energy source significantly affect the number and function of ILC2s in addition to affecting their interactions with resident cell types. How ILC2s react to changes in the metabolic environment is a clear determinant of the severity of disease. Treating sources of metabolic instability via critical immune cells provides a clear avenue for modulation of systemic homeostasis and new treatments of T2DM.
Collapse
Affiliation(s)
- Jacob D Painter
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
5
|
Li H, Hostager BS, Arkee T, Bishop GA. Multiple mechanisms for TRAF3-mediated regulation of the T cell costimulatory receptor GITR. J Biol Chem 2021; 297:101097. [PMID: 34418432 PMCID: PMC8441216 DOI: 10.1016/j.jbc.2021.101097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 11/28/2022] Open
Abstract
Tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) plays context-specific roles in multiple receptor-mediated signaling pathways in different cell types. Mice lacking TRAF3 in T cells display defective T-cell-mediated immune responses to immunization and infection and demonstrate defective early signaling via the TCR complex. However, the role of TRAF3 in the function of GITR/TNFRSF18, an important costimulatory member of the TNFR superfamily, is unclear. Here we investigated the impact of T cell TRAF3 status on both GITR expression and activation of specific kinases in the GITR signaling pathway in T cells. Our results indicate that TRAF3 negatively regulates GITR functions in several ways. First, expression of GITR protein was elevated in TRAF3-deficient T cells, resulting from both transcriptional and posttranslational regulation that led to greater GITR transcript levels, as well as enhanced GITR protein stability. TRAF3 associated with T cell GITR in a manner dependent upon GITR ligation. TRAF3 also inhibited several events of the GITR mediated early signaling cascade, in a manner independent of recruitment of phosphatases, a mechanism by which TRAF3 inhibits signaling through several other cytokine receptors. These results add new information to our understanding of GITR signaling and function in T cells, which is relevant to the potential use of GITR to enhance immune therapies.
Collapse
Affiliation(s)
- Hanzeng Li
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, Iowa, USA
| | - Bruce S Hostager
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, Iowa, USA
| | - Tina Arkee
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, Iowa, USA; Medical Scientist Training Program, The University of Iowa, Iowa City, Iowa, USA
| | - Gail A Bishop
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, Iowa, USA; Medical Scientist Training Program, The University of Iowa, Iowa City, Iowa, USA; Department of Internal Medicine, The University of Iowa, Iowa City, Iowa, USA; Research, Iowa City VA Medical Center, Iowa City, Iowa, USA.
| |
Collapse
|
6
|
Kelly WJ, Giles AJ, Gilbert M. T lymphocyte-targeted immune checkpoint modulation in glioma. J Immunother Cancer 2021; 8:jitc-2019-000379. [PMID: 32051289 PMCID: PMC7057419 DOI: 10.1136/jitc-2019-000379] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2020] [Indexed: 02/07/2023] Open
Abstract
Immunomodulatory therapies targeting inhibitory checkpoint molecules have revolutionized the treatment of solid tumor malignancies. Concerns about whether systemic administration of an immune checkpoint inhibitor could impact primary brain tumors were answered with the observation of definitive responses in pediatric patients harboring hypermutated gliomas. Although initial clinical results in patients with glioblastoma (GBM) were disappointing, recently published results have demonstrated a potential survival benefit in patients with recurrent GBM treated with neoadjuvant programmed cell death protein 1 blockade. While these findings necessitate verification in subsequent studies, they support the possibility of achieving clinical meaningful immune responses in malignant primary brain tumors including GBM, a disease in dire need of additional therapeutic options. There are several challenges involved in treating glioma with immune checkpoint modulators including the immunosuppressive nature of GBM itself with high inhibitory checkpoint expression, the immunoselective blood brain barrier impairing the ability for peripheral lymphocytes to traffic to the tumor microenvironment and the high prevalence of corticosteroid use which suppress lymphocyte activation. However, by simultaneously targeting multiple costimulatory and inhibitory pathways, it may be possible to achieve an effective antitumoral immune response. To this end, there are now several novel agents targeting more recently uncovered “second generation” checkpoint molecules. Given the multiplicity of drugs being considered for combination regimens, an increased understanding of the mechanisms of action and resistance combined with more robust preclinical and early clinical testing will be needed to be able to adequately test these agents. This review summarizes our current understanding of T lymphocyte-modulating checkpoint molecules as it pertains to glioma with the hope for a renewed focus on the most promising therapeutic strategies.
Collapse
Affiliation(s)
| | - Amber Jin Giles
- Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark Gilbert
- Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
7
|
Pattarabanjird T, Li C, McNamara C. B Cells in Atherosclerosis: Mechanisms and Potential Clinical Applications. ACTA ACUST UNITED AC 2021; 6:546-563. [PMID: 34222726 PMCID: PMC8246059 DOI: 10.1016/j.jacbts.2021.01.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/05/2021] [Accepted: 01/05/2021] [Indexed: 12/17/2022]
Abstract
B cells regulate atherosclerotic plaque formation through production of antibodies and cytokines, and effects are subset specific (B1 and B2). Putative human atheroprotective B1 cells function similarly to murine B1 in their spontaneous IgM antibody production. However, marker strategies in identifying human and murine B1 are different. IgM antibody to oxidation specific epitopes produced by B1 cells associate with human coronary artery disease. Neoantigen immunization may be a promising strategy for atherosclerosis vaccine development, but further study to determine relevant antigens still need to be done. B-cell–targeted therapies, used in treating autoimmune diseases as well as lymphoid cancers, might have potential applications in treating cardiovascular diseases. Short- and long-term cardiovascular effects of these agents need to be assessed.
Because atherosclerotic cardiovascular disease is a leading cause of death worldwide, understanding inflammatory processes underpinning its pathology is critical. B cells have been implicated as a key immune cell type in regulating atherosclerosis. B-cell effects, mediated by antibodies and cytokines, are subset specific. In this review, we focus on elaborating mechanisms underlying subtype-specific roles of B cells in atherosclerosis and discuss available human data implicating B cells in atherosclerosis. We further discuss potential B cell–linked therapeutic approaches, including immunization and B cell–targeted biologics. Given recent evidence strongly supporting a role for B cells in human atherosclerosis and the expansion of immunomodulatory agents that affect B-cell biology in clinical use and clinical trials for other disorders, it is important that the cardiovascular field be cognizant of potential beneficial or untoward effects of modulating B-cell activity on atherosclerosis.
Collapse
Key Words
- APRIL, A proliferation−inducing ligand
- ApoE, apolipoprotein E
- B-cell
- BAFF, B-cell–activating factor
- BAFFR, B-cell–activating factor receptor
- BCMA, B-cell maturation antigen
- BCR, B-cell receptor
- Breg, regulatory B cell
- CAD, coronary artery disease
- CTLA4, cytotoxic T-lymphocyte–associated protein 4
- CVD, cardiovascular disease
- CXCR4, C-X-C motif chemokine receptor 4
- GC, germinal center
- GITR, glucocorticoid-induced tumor necrosis factor receptor–related protein
- GITRL, glucocorticoid-induced tumor necrosis factor receptor–related protein ligand
- GM-CSF, granulocyte-macrophage colony–stimulating factor
- ICI, immune checkpoint inhibitor
- IFN, interferon
- IL, interleukin
- IVUS, intravascular ultrasound
- LDL, low-density lipoprotein
- LDLR, low-density lipoprotein receptor
- MDA-LDL, malondialdehyde-modified low-density lipoprotein
- MI, myocardial infarction
- OSE, oxidation-specific epitope
- OxLDL, oxidized low-density lipoprotein
- PC, phosphorylcholine
- PD-1, programmed cell death protein 1
- PD-L2, programmed death ligand 2
- PDL1, programmed death ligand 1
- RA, rheumatoid arthritis
- SLE, systemic lupus erythematosus
- TACI, transmembrane activator and CAML interactor
- TNF, tumor necrosis factor
- Treg, regulatory T cell
- atherosclerosis
- immunoglobulins
- mAb, monoclonal antibody
Collapse
Affiliation(s)
- Tanyaporn Pattarabanjird
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA.,Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Cynthia Li
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Coleen McNamara
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA.,Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
8
|
Chen B, Yang M, Li K, Li J, Xu L, Xu F, Xu Y, Ren D, Zhang J, Liu L. Immune-related genes and gene sets for predicting the response to anti-programmed death 1 therapy in patients with primary or metastatic non-small cell lung cancer. Oncol Lett 2021; 22:540. [PMID: 34084219 PMCID: PMC8161458 DOI: 10.3892/ol.2021.12801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 04/14/2021] [Indexed: 12/24/2022] Open
Abstract
Although antibodies targeting the immune checkpoint protein programmed death-1 (PD-1) exert therapeutic effects in patients with primary or metastatic non-small cell lung cancer (NSCLC), the majority of patients exhibit partial or complete resistance to anti-PD1 treatment. Thus, the aim of the present study was to identify reliable biomarkers for predicting the response to anti-PD-1 therapy. The present study analyzed tumor specimens isolated from 24 patients (13 with primary and 11 with metastatic NSCLC) prior to treatment with approved PD1-targeting antibodies. The expression profile of 395 immune-related genes was examined using RNA immune-oncology panel sequencing. The results demonstrated that six immune-related differently expressed genes (DEGs), including HLA-F-AS1, NCF1, RORC, DMBT1, KLRF1 and IL-18, and five DEGs, including HLA-A, HLA-DPA1, TNFSF18, IFI6 and PTK7, may be used as single biomarkers for predicting the efficacy of anti-PD-1 treatment in patients with primary and with metastatic NSCLC, respectively. In addition, two DEG sets comprising either six (HLA-F-AS1, NCF1, RORC, DMBT1, KLRF and IL-18) or two (HLA-A and TNFSF18) DEGs as potential combination biomarkers for predicting the efficacy of anti-PD-1 therapy in patients with NSCLC. Patients with a calculated expression level of the DEG sets >6.501 (primary NSCLC) or >6.741 (metastatic NSCLC) may benefit from the anti-PD-1 therapy. Overall, these findings provided a basis for the identification of additional biomarkers for predicting the response to anti-PD-1 treatment.
Collapse
Affiliation(s)
- Bolin Chen
- Thoracic Medicine Department 2, Cancer Hospital Affiliated to Xiangya Medical College, Central South University, Changsha, Hunan 410013, P.R. China
| | - Min Yang
- Department of Respiratory Disease, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Kang Li
- Thoracic Medicine Department 2, Cancer Hospital Affiliated to Xiangya Medical College, Central South University, Changsha, Hunan 410013, P.R. China
| | - Jia Li
- Thoracic Medicine Department 2, Cancer Hospital Affiliated to Xiangya Medical College, Central South University, Changsha, Hunan 410013, P.R. China
| | - Li Xu
- Thoracic Medicine Department 2, Cancer Hospital Affiliated to Xiangya Medical College, Central South University, Changsha, Hunan 410013, P.R. China
| | - Fang Xu
- Thoracic Medicine Department 2, Cancer Hospital Affiliated to Xiangya Medical College, Central South University, Changsha, Hunan 410013, P.R. China
| | - Yan Xu
- Thoracic Medicine Department 2, Cancer Hospital Affiliated to Xiangya Medical College, Central South University, Changsha, Hunan 410013, P.R. China
| | - Dandan Ren
- Genecast Biotechnology Co., Ltd., Beijing 100089, P.R. China
| | - Jiao Zhang
- Genecast Biotechnology Co., Ltd., Beijing 100089, P.R. China
| | - Liyu Liu
- Thoracic Medicine Department 2, Cancer Hospital Affiliated to Xiangya Medical College, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
9
|
Shami A, Atzler D, Bosmans LA, Winkels H, Meiler S, Lacy M, van Tiel C, Ta Megens R, Nitz K, Baardman J, Kusters P, Seijkens T, Buerger C, Janjic A, Riccardi C, Edsfeldt A, Monaco C, Daemen M, de Winther MPJ, Nilsson J, Weber C, Gerdes N, Gonçalves I, Lutgens E. Glucocorticoid-induced tumour necrosis factor receptor family-related protein (GITR) drives atherosclerosis in mice and is associated with an unstable plaque phenotype and cerebrovascular events in humans. Eur Heart J 2021; 41:2938-2948. [PMID: 32728688 DOI: 10.1093/eurheartj/ehaa484] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/21/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022] Open
Abstract
AIMS GITR-a co-stimulatory immune checkpoint protein-is known for both its activating and regulating effects on T-cells. As atherosclerosis bears features of chronic inflammation and autoimmunity, we investigated the relevance of GITR in cardiovascular disease (CVD). METHODS AND RESULTS GITR expression was elevated in carotid endarterectomy specimens obtained from patients with cerebrovascular events (n = 100) compared to asymptomatic patients (n = 93) and correlated with parameters of plaque vulnerability, including plaque macrophage, lipid and glycophorin A content, and levels of interleukin (IL)-6, IL-12, and C-C-chemokine ligand 2. Soluble GITR levels were elevated in plasma from subjects with CVD compared to healthy controls. Plaque area in 28-week-old Gitr-/-Apoe-/- mice was reduced, and plaques had a favourable phenotype with less macrophages, a smaller necrotic core and a thicker fibrous cap. GITR deficiency did not affect the lymphoid population. RNA sequencing of Gitr-/-Apoe-/- and Apoe-/- monocytes and macrophages revealed altered pathways of cell migration, activation, and mitochondrial function. Indeed, Gitr-/-Apoe-/- monocytes displayed decreased integrin levels, reduced recruitment to endothelium, and produced less reactive oxygen species. Likewise, GITR-deficient macrophages produced less cytokines and had a reduced migratory capacity. CONCLUSION Our data reveal a novel role for the immune checkpoint GITR in driving myeloid cell recruitment and activation in atherosclerosis, thereby inducing plaque growth and vulnerability. In humans, elevated GITR expression in carotid plaques is associated with a vulnerable plaque phenotype and adverse cerebrovascular events. GITR has the potential to become a novel therapeutic target in atherosclerosis as it reduces myeloid cell recruitment to the arterial wall and impedes atherosclerosis progression.
Collapse
Affiliation(s)
- Annelie Shami
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany.,Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians Universität, München, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Laura A Bosmans
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Holger Winkels
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany.,Department of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Svenja Meiler
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany
| | - Michael Lacy
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Claudia van Tiel
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Remco Ta Megens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany.,Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
| | - Katrin Nitz
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany
| | - Jeroen Baardman
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Pascal Kusters
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Tom Seijkens
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Christina Buerger
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany
| | - Aleksandar Janjic
- Anthropology & Human Genomics, Department of Biology II, Ludwig-Maximilians-Universität, München, Martinsried, Germany
| | - Carlo Riccardi
- Department of Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Andreas Edsfeldt
- Department of Clinical Sciences Malmö, Lund University, Clinical Research Center, Malmö, Sweden.,Department of Cardiology, Skåne University Hospital, Lund University, Sweden
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, UK
| | - Mat Daemen
- Department of Pathology, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Menno P J de Winther
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Clinical Research Center, Malmö, Sweden
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Isabel Gonçalves
- Department of Clinical Sciences Malmö, Lund University, Clinical Research Center, Malmö, Sweden.,Department of Cardiology, Skåne University Hospital, Lund University, Sweden
| | - Esther Lutgens
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
10
|
Wang Y, Liao K, Liu B, Niu C, Zou W, Yang L, Wang T, Tian D, Luo Z, Dai J, Li Q, Liu E, Gong C, Fu Z, Li Y, Ding F. GITRL on dendritic cells aggravates house dust mite-induced airway inflammation and airway hyperresponsiveness by modulating CD4 + T cell differentiation. Respir Res 2021; 22:46. [PMID: 33557842 PMCID: PMC7869253 DOI: 10.1186/s12931-020-01583-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/22/2020] [Indexed: 01/01/2023] Open
Abstract
Background Glucocorticoid-induced tumor necrosis factor receptor family-related protein ligand (GITRL) plays an important role in tumors, autoimmunity and inflammation. However, GITRL is not known to modulate the pathogenesis of allergic asthma. In this study, we investigated whether regulating GITRL expressed on dendritic cells (DCs) can prevent asthma and to elucidate its mechanism of action. Methods In vivo, the role of GITRL in modulating house dust mite (HDM)-induced asthma was assessed in adeno-associated virus (AAV)-shGITRL mice. In vitro, the role of GITRL expression by DCs was evaluated in LV-shGITRL bone marrow dendritic cells (BMDCs) under HDM stimulation. And the direct effect of GITRL was observed by stimulating splenocytes with GITRL protein. The effect of regulating GITRL on CD4+ T cell differentiation was detected. Further, GITRL mRNA in the peripheral blood of asthmatic children was tested. Results GITRL was significantly increased in HDM-challenged mice. In GITRL knockdown mice, allergen-induced airway inflammation, serum total IgE levels and airway hyperresponsiveness (AHR) were reduced. In vitro, GITRL expression on BMDCs was increased after HDM stimulation. Further, knocking down GITRL on DCs partially restored the balance of Th1/Th2 and Th17/Treg cells. Moreover, GITRL stimulation in vitro inhibited Treg cell differentiation and promoted Th2 and Th17 cell differentiation. Similarly, GITRL mRNA expression was increased in the peripheral blood from asthmatic children. Conclusions This study identified a novel role for GITRL expressed by DCs as a positive regulator of CD4+ T cells responses in asthma, which implicates that GITRL inhibitors may be a potential immunotherapy for asthma.
Collapse
Affiliation(s)
- Yaping Wang
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Kou Liao
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Bo Liu
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Department of Cardiothoracic Surgery, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Chao Niu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Wenjing Zou
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Lili Yang
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Ting Wang
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Daiyin Tian
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhengxiu Luo
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jihong Dai
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Qubei Li
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Enmei Liu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Caihui Gong
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhou Fu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Ying Li
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China. .,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Fengxia Ding
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Yuzhong District, No. 136, Zhongshan 2nd Road, Chongqing, 400014, China. .,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
11
|
Geva R, Voskoboynik M, Dobrenkov K, Mayawala K, Gwo J, Wnek R, Chartash E, Long GV. First-in-human phase 1 study of MK-1248, an anti-glucocorticoid-induced tumor necrosis factor receptor agonist monoclonal antibody, as monotherapy or with pembrolizumab in patients with advanced solid tumors. Cancer 2020; 126:4926-4935. [PMID: 32809217 DOI: 10.1002/cncr.33133] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/25/2020] [Accepted: 07/02/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Ligation of glucocorticoid-induced tumor necrosis factor receptor (GITR) decreases regulatory T cell-mediated suppression and enhances T-cell proliferation, effector function, and survival. MK-1248 is a humanized immunoglobulin G4 anti-GITR monoclonal antibody agonist. METHODS In patients with advanced solid tumors, MK-1248 (starting dose, 0.12 mg) was tested alone and with pembrolizumab (200 mg) according to a 3 + 3 dose escalation design (ClinicalTrials.gov identifier NCT02553499); both treatments were administered intravenously every 3 weeks for ≤4 and ≤35 cycles, respectively. The safety and tolerability, maximum tolerated dose, and pharmacokinetics/pharmacodynamics were explored. RESULTS Twenty patients received MK-1248 monotherapy; 17 received combination therapy. The most frequent tumor types were colorectal cancer (n = 8), melanoma (n = 6), and renal cell carcinoma (n = 4). MK-1248 was generally well tolerated at the maximum tested doses of 170 (monotherapy) and 60 mg (combination). No dose-limiting toxicities (DLTs) or treatment-related deaths occurred. Adverse events (AEs) occurred in 36 of the 37 patients (97%); the most common were vomiting (n = 13 [35%]), anemia (n = 10 [27%]), and decreased appetite (n = 10 [27%]). Grade 3 to 5 AEs occurred in 19 of the 37 patients (51%). Treatment-related AEs occurred in 18 of the 37 patients (49%): 9 of the 20 patients (45%) on monotherapy and 9 of the 17 patients (53%) on combination therapy. Among the 17 patients receiving combination therapy, 1 achieved a complete response and 2 achieved a partial response, for an objective response rate of 18%; no patients achieved an objective response with monotherapy. The disease control rate (stable disease or better) was 15% with monotherapy and 41% with combination therapy. CONCLUSIONS MK-1248 was generally well tolerated at doses up to 170 (monotherapy) and 60 mg (combination), with no DLTs or treatment-related deaths. Combination therapy provided limited antitumor responses.
Collapse
Affiliation(s)
- Ravit Geva
- Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Mark Voskoboynik
- Nucleus Network, Alfred Hospital, Monash University, Melbourne, Victoria, Australia
| | | | | | | | | | | | - Georgina V Long
- Royal North Shore Hospital, St. Leonards, New South Wales, Australia.,Melanoma Institute Australia, Wollstonecraft, New South Wales, Australia.,The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
12
|
Ho LP, Teo Bryan YW, Linn YC, Tan PH, Jabed I, Toh HC, Yeoh KW. Unifying framework for acute resolution, malignancy and autoimmunity. Scand J Immunol 2020; 91:e12869. [PMID: 32017146 DOI: 10.1111/sji.12869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/27/2020] [Accepted: 02/01/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Liam Pock Ho
- Department of Clinical Pathology, Singapore General Hospital, Singapore, Singapore.,Department of Hematology, Singapore General Hospital, Singapore, Singapore
| | - Yi Wei Teo Bryan
- Department of Clinical Research, Singapore General Hospital, Singapore, Singapore
| | - Yeh Ching Linn
- Department of Hematology, Singapore General Hospital, Singapore, Singapore
| | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Iqbal Jabed
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Han Chong Toh
- Department of Medical Oncology, National Cancer Center, Singapore, Singapore
| | - Kheng Wei Yeoh
- Department of Radiation Oncology, National Cancer Center, Singapore, Singapore
| |
Collapse
|
13
|
Abstract
The recent clinical success of cancer immunotherapy with checkpoint blockade has led to renewed interest into the development of immune modulatory agents with the capacity to activate anti-tumor T cell responses. Standardization of optimized in vitro assays for efficient assessment of immune function of such new drugs is thus needed to facilitate clinical development of the optimal drug candidates. Here, we describe an optimized version of T cell suppression assay designed to test the effect of immunomodulatory agents on T cell function and activation. We apply this assay to investigate the agonist activity of the T cell co-stimulatory molecule glucocorticoid-induced TNFR-related protein (GITR). We detail a protocol for concurrent assessment of multiple levels of T cell functional modulation upon GITR engagement, including T cell priming, activation and effector function, in a single assay. As human GITR agonist antibodies are currently under development, availability of standardized cell-based functional assays of GITR agonism is instrumental to translate anti-GITR therapy into the clinical setting.
Collapse
|
14
|
Vence L, Bucktrout SL, Fernandez Curbelo I, Blando J, Smith BM, Mahne AE, Lin JC, Park T, Pascua E, Sai T, Chaparro-Riggers J, Subudhi SK, Scutti JB, Higa MG, Zhao H, Yadav SS, Maitra A, Wistuba II, Allison JP, Sharma P. Characterization and Comparison of GITR Expression in Solid Tumors. Clin Cancer Res 2019; 25:6501-6510. [PMID: 31358539 DOI: 10.1158/1078-0432.ccr-19-0289] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/16/2019] [Accepted: 07/11/2019] [Indexed: 12/19/2022]
Abstract
PURPOSE Determine the differential effect of a FcγR-binding, mIgG2a anti-GITR antibody in mouse tumor models, and characterize the tumor microenvironment for the frequency of GITR expression in T-cell subsets from seven different human solid tumors.Experimental Design: For mouse experiments, wild-type C57BL/6 mice were subcutaneously injected with MC38 cells or B16 cells, and BALB/c mice were injected with CT26 cells. Mice were treated with the anti-mouse GITR agonist antibody 21B6, and tumor burden and survival were monitored. GITR expression was evaluated at the single-cell level using flow cytometry (FC). A total of 213 samples were evaluated for GITR expression by IHC, 63 by FC, and 170 by both in seven human solid tumors: advanced hepatocellular carcinoma, non-small cell lung cancer (NSCLC), renal cell carcinoma, pancreatic carcinoma, head and neck carcinoma, melanoma, and ovarian carcinoma. RESULTS The therapeutic benefit of 21B6 was greatest in CT26 followed by MC38, and was least in the B16 tumor model. The frequency of CD8 T cells and effector CD4 T cells within the immune infiltrate correlated with response to treatment with GITR antibody. Analysis of clinical tumor samples showed that NSCLC, renal cell carcinoma, and melanoma had the highest proportions of GITR-expressing cells and highest per-cell density of GITR expression on CD4+ Foxp3+ T regulatory cells. IHC and FC data showed similar trends with a good correlation between both techniques. CONCLUSIONS Human tumor data suggest that NSCLC, renal cell carcinoma, and melanoma should be the tumor subtypes prioritized for anti-GITR therapy development.
Collapse
Affiliation(s)
- Luis Vence
- The Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samantha L Bucktrout
- Cancer Immunology Discovery Unit, South San Francisco, California.,Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Irina Fernandez Curbelo
- The Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorge Blando
- The Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bevin M Smith
- Cancer Immunology Discovery Unit, South San Francisco, California
| | - Ashley E Mahne
- Cancer Immunology Discovery Unit, South San Francisco, California
| | - John C Lin
- Cancer Immunology Discovery Unit, South San Francisco, California.,Regeneron Pharmaceuticals Inc., Tarrytown, New York
| | - Terrence Park
- Cancer Immunology Discovery Unit, South San Francisco, California
| | - Edward Pascua
- Cancer Immunology Discovery Unit, South San Francisco, California
| | - Tao Sai
- Cancer Immunology Discovery Unit, South San Francisco, California
| | | | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorge B Scutti
- The Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria G Higa
- The Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hao Zhao
- The Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shalini S Yadav
- The Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James P Allison
- The Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Padmanee Sharma
- The Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
15
|
Richards DM, Marschall V, Billian-Frey K, Heinonen K, Merz C, Redondo Müller M, Sefrin JP, Schröder M, Sykora J, Fricke H, Hill O, Gieffers C, Thiemann M. HERA-GITRL activates T cells and promotes anti-tumor efficacy independent of FcγR-binding functionality. J Immunother Cancer 2019; 7:191. [PMID: 31324216 PMCID: PMC6642547 DOI: 10.1186/s40425-019-0671-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/08/2019] [Indexed: 11/28/2022] Open
Abstract
Background Glucocorticoid-induced TNFR-related protein (TNFRSF18, GITR, CD357), expressed by T cells, and its ligand (TNFSF18, GITRL), expressed by myeloid populations, provide co-stimulatory signals that boost T cell activity. Due to the important role that GITR plays in regulating immune functions, agonistic stimulation of GITR is a promising therapeutic concept. Multiple strategies to induce GITR signaling have been investigated. The limited clinical efficacy of antibody-based GITR agonists results from structural and functional characteristics of antibodies that are unsuitable for stimulating the well-defined trimeric members of the TNFRSF. Methods To overcome limitations of antibody-based TNFRSF agonists, we have developed HERA-GITRL, a fully human hexavalent TNF receptor agonist (HERA) targeting GITR and mimicking the natural signaling concept. HERA-GITRL is composed of a trivalent but single-chain GITRL-receptor-binding-domain (scGITRL-RBD) unit fused to an IgG1 derived silenced Fc-domain serving as dimerization scaffold. A specific mouse surrogate, mmHERA-GITRL, was also generated to examine in vivo activity in respective mouse tumor models. Results For functional characterization of HERA-GITRL in vitro, human immune cells were isolated from healthy-donor blood and stimulated with anti-CD3 antibody in the presence of HERA-GITRL. Consistently, HERA-GITRL increased the activity of T cells, including proliferation and differentiation, even in the presence of regulatory T cells. In line with these findings, mmHERA-GITRL enhanced antigen-specific clonal expansion of both CD4+ (OT-II) and CD8+ (OT-I) T cells in vivo while having no effect on non-specific T cells. In addition, mmHERA-GITRL showed single-agent anti-tumor activity in two subcutaneous syngeneic colon cancer models (CT26wt and MC38-CEA). Importantly, this activity is independent of its FcγR-binding functionality, as both mmHERA-GITRL with a functional Fc- and a silenced Fc-domain showed similar tumor growth inhibition. Finally, in a direct in vitro comparison to a bivalent clinical benchmark anti-GITR antibody and a trivalent GITRL, only the hexavalent HERA-GITRL showed full biological activity independent of additional crosslinking. Conclusion In this manuscript, we describe the development of HERA-GITRL, a true GITR agonist with a clearly defined mechanism of action. By clustering six receptor chains in a spatially well-defined manner, HERA-GITRL induces potent agonistic activity without being dependent on additional FcγR-mediated crosslinking. Electronic supplementary material The online version of this article (10.1186/s40425-019-0671-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David M Richards
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | | | - Katharina Billian-Frey
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Karl Heinonen
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Christian Merz
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | | | - Julian P Sefrin
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Matthias Schröder
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Jaromir Sykora
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | | | - Oliver Hill
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Christian Gieffers
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Meinolf Thiemann
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany.
| |
Collapse
|
16
|
Li Y, Yang S, Li Z, Meng H, Jin W, Yang H, Yin W. Soluble glucocorticoid-induced tumor necrosis factor receptor regulates Helios expression in myasthenia gravis. J Transl Med 2019; 17:168. [PMID: 31118027 PMCID: PMC6530055 DOI: 10.1186/s12967-019-1916-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 05/10/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Helios is important for functional and phenotype stability of regulatory T cells (Tregs). However, the role of Helios in autoimmune diseases and its regulation remains unclear. This study aimed to investigate the role of Helios+ Tregs in myasthenia gravis (MG) and glucocorticoid-induced tumor necrosis factor receptor (GITR) and its ligand (GITRL) in the modulation of Helios. METHOD Multicolor flow cytometry was performed to analyze Helios+ Tregs in peripheral blood from MG patients and healthy donors (HDs). Enzyme-linked immunosorbent assay (ELISA) was used to determine the levels of soluble GITRL/GITR in plasma. Tregs were isolated via magnetic separation and treated with recombinant GITRL and GITR-Fc. Membrane GITRL on Tregs and expression of Helios and other markers (FOXP3, CD25, CD39, CTLA-4, PD-L1 and IL-10) involved in immunosuppressive activity were determined by flow cytometry. RESULT Both Helios+ Tregs and soluble GITR were decreased in generalized MG (GMG) patients (n = 14), compared with HDs (n = 14) and ocular MG (OMG) patients (n = 16). Helios+ Tregs possessed greater immunosuppressive capacity compared to Helios- Tregs. Further analysis indicates soluble GITR was negatively correlated with quantitative MG score and promoted Helios expression and enhanced function of Tregs independently of membrane GITRL. CONCLUSION This work demonstrates abnormal changes in Helios+ Tregs and soluble GITR in MG, as well as direct regulation of Helios by GITR in the context of Tregs. This work provides new insight into the role of GITR in the regulatory pathway of Helios and pathogenesis of MG.
Collapse
Affiliation(s)
- Yi Li
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Shumei Yang
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Zhibin Li
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Huanyu Meng
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Wanling Jin
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China.
| | - Weifan Yin
- Department of Neurology, Second Xiangya Hospital, Central South University, 137 People Road, Changsha, Hunan, China.
| |
Collapse
|
17
|
Zappasodi R, Sirard C, Li Y, Budhu S, Abu-Akeel M, Liu C, Yang X, Zhong H, Newman W, Qi J, Wong P, Schaer D, Koon H, Velcheti V, Hellmann MD, Postow MA, Callahan MK, Wolchok JD, Merghoub T. Rational design of anti-GITR-based combination immunotherapy. Nat Med 2019; 25:759-766. [PMID: 31036879 PMCID: PMC7457830 DOI: 10.1038/s41591-019-0420-8] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/12/2019] [Indexed: 12/13/2022]
Abstract
Modulating T cell homeostatic mechanisms with checkpoint blockade can efficiently promote endogenous anti-tumor T cell responses1-11. However, many patients still do not benefit from checkpoint blockade12, highlighting the need for targeting of alternative immune pathways13. Glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) is an attractive target for immunotherapy, owing to its capacity to promote effector T cell (Teff) functions14,15 and hamper regulatory T cell (Treg) suppression16-20. On the basis of the potent preclinical anti-tumor activity of agonist anti-GITR antibodies, reported by us and others16,21,22, we initiated the first in-human phase 1 trial of GITR agonism with the anti-GITR antibody TRX518 ( NCT01239134 ). Here, we report the safety profile and immune effects of TRX518 monotherapy in patients with advanced cancer and provide mechanistic preclinical evidence to rationally combine GITR agonism with checkpoint blockade in future clinical trials. We demonstrate that TRX518 reduces circulating and intratumoral Treg cells to similar extents, providing an easily assessable biomarker of anti-GITR activity. Despite Treg reductions and increased Teff:Treg ratios, substantial clinical responses were not seen. Similarly, in mice with advanced tumors, GITR agonism was not sufficient to activate cytolytic T cells due to persistent exhaustion. We demonstrate that T cell reinvigoration with PD-1 blockade can overcome resistance of advanced tumors to anti-GITR monotherapy. These findings led us to start investigating TRX518 with PD-1 pathway blockade in patients with advanced refractory tumors ( NCT02628574 ).
Collapse
Affiliation(s)
- Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Yanyun Li
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sadna Budhu
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mohsen Abu-Akeel
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cailian Liu
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xia Yang
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hong Zhong
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jingjing Qi
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immune Monitoring Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Phillip Wong
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immune Monitoring Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Schaer
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Henry Koon
- Case Western Reserve University, Cleveland, OH, USA
| | - Vamsidhar Velcheti
- Department of Hematology and Oncology, New York University School of Medicine, New York, NY, USA
| | - Matthew D Hellmann
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Michael A Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Margaret K Callahan
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Jedd D Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
18
|
Lee WH, Seo D, Lim SG, Suk K. Reverse Signaling of Tumor Necrosis Factor Superfamily Proteins in Macrophages and Microglia: Superfamily Portrait in the Neuroimmune Interface. Front Immunol 2019; 10:262. [PMID: 30838001 PMCID: PMC6389649 DOI: 10.3389/fimmu.2019.00262] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 01/30/2019] [Indexed: 12/14/2022] Open
Abstract
The tumor necrosis factor (TNF) superfamily (TNFSF) is a protein superfamily of type II transmembrane proteins commonly containing the TNF homology domain. The superfamily contains more than 20 protein members, which can be released from the cell membrane by proteolytic cleavage. Members of the TNFSF function as cytokines and regulate diverse biological processes, including immune responses, proliferation, differentiation, apoptosis, and embryogenesis, by binding to TNFSF receptors. Many TNFSF proteins are also known to be responsible for the regulation of innate immunity and inflammation. Both receptor-mediated forward signaling and ligand-mediated reverse signaling play important roles in these processes. In this review, we discuss the functional expression and roles of various reverse signaling molecules and pathways of TNFSF members in macrophages and microglia in the central nervous system (CNS). A thorough understanding of the roles of TNFSF ligands and receptors in the activation of macrophages and microglia may improve the treatment of inflammatory diseases in the brain and periphery. In particular, TNFSF reverse signaling in microglia can be exploited to gain further insights into the functions of the neuroimmune interface in physiological and pathological processes in the CNS.
Collapse
Affiliation(s)
- Won-Ha Lee
- BK21 Plus KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, South Korea
| | - Donggun Seo
- BK21 Plus KNU Biomedical Convergence Program, Department of Pharmacology, School of Medicine, Brain Science & Engineering Institute, Kyungpook National University, Daegu, South Korea
| | - Su-Geun Lim
- BK21 Plus KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, South Korea
| | - Kyoungho Suk
- BK21 Plus KNU Biomedical Convergence Program, Department of Pharmacology, School of Medicine, Brain Science & Engineering Institute, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
19
|
Immune checkpoint blockade and its combination therapy with small-molecule inhibitors for cancer treatment. Biochim Biophys Acta Rev Cancer 2018; 1871:199-224. [PMID: 30605718 DOI: 10.1016/j.bbcan.2018.12.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 02/05/2023]
Abstract
Initially understood for its physiological maintenance of self-tolerance, the immune checkpoint molecule has recently been recognized as a promising anti-cancer target. There has been considerable interest in the biology and the action mechanism of the immune checkpoint therapy, and their incorporation with other therapeutic regimens. Recently the small-molecule inhibitor (SMI) has been identified as an attractive combination partner for immune checkpoint inhibitors (ICIs) and is becoming a novel direction for the field of combination drug design. In this review, we provide a systematic discussion of the biology and function of major immune checkpoint molecules, and their interactions with corresponding targeting agents. With both preclinical studies and clinical trials, we especially highlight the ICI + SMI combination, with its recent advances as well as its application challenges.
Collapse
|
20
|
Khalife E, Khodadadi A, Talaeizadeh A, Rahimian L, Nemati M, Jafarzadeh A. Overexpression of Regulatory T Cell-Related Markers (FOXP3, CTLA-4 and GITR) by Peripheral Blood Mononuclear Cells from Patients with Breast Cancer. Asian Pac J Cancer Prev 2018; 19:3019-3025. [PMID: 30484986 PMCID: PMC6318404 DOI: 10.31557/apjcp.2018.19.11.3019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background: Regulatory T (Treg) cells are immunosuppressor lymphocytes that play a critical role in the establishment and progression of cancers. A number of markers, especially FOXP3, CTLA-4 and GITR influence the function of Treg cells. This investigation aimed to evaluate the expression of a number of important Treg cell-related markers by peripheral blood mononuclear cells (PBMCs) from newly-diagnosed women with breast cancer. Methods: The fresh PBMCs were obtained from 20 women with breast cancer and 20 healthy individuals. The PBMCs from both groups were cultured for 32 hours in the presence or absence of PHA (10 μg/ml). After total RNA extraction from cultured PBMCs, the expression of the FOXP3, CTLA-4 and GITR transcripts was assessed using real time-PCR. Results: The mRNA expression of FOXP3, CTLA-4 and GITR in unstimulated PBMCs from patients with breast cancer were significantly higher than healthy control group (P<0.05, P<0.03 and P<0.04, respectively). Similarly, the expression of FOXP3, CTLA-4 and GITR transcripts in PHA-stimulated PBMCs from patients with breast cancer were significantly increased in comparison with healthy individuals (P<0.01, P<0.005 and P<0.01, respectively). Conclusion: The increased expression of FOXP3, CTLA-4 and GITR represent higher activity of Treg cells in patients with breast cancer that may play an important role in the tumor establishment and development.
Collapse
Affiliation(s)
- Esmat Khalife
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | | | | | | | | | | |
Collapse
|
21
|
Kleef R, Moss R, Szasz AM, Bohdjalian A, Bojar H, Bakacs T. Complete Clinical Remission of Stage IV Triple-Negative Breast Cancer Lung Metastasis Administering Low-Dose Immune Checkpoint Blockade in Combination With Hyperthermia and Interleukin-2. Integr Cancer Ther 2018; 17:1297-1303. [PMID: 30193538 PMCID: PMC6247552 DOI: 10.1177/1534735418794867] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The prognosis of triple-negative breast cancer with metastases after chemotherapy
remains dismal. We report the case of a 50-year-old female with first disease
recurrence at the axillary lymph node and, later on, bilateral pulmonary
metastases with severe shortness of breath. The patient received low-dose immune
checkpoint blockade (concurrent nivolumab and ipilimumab) weekly over 3 weeks
with regional hyperthermia 3 times a week, followed by systemic fever-range
hyperthermia induced by interleukin-2 for 5 days. She went into complete
remission of her pulmonary metastases with transient WHO I-II diarrhea and skin
rash. The patient remained alive for 27 months after the start of treatment,
with recurrence of metastases as a sternal mass, and up to 3 cm pleural
metastases. This exceptional response should instigate further research efforts
with this protocol, which consists only of approved drugs and treatments.
Collapse
Affiliation(s)
- Ralf Kleef
- 1 Immunology & Integrative Oncology, Vienna, Austria
| | | | - A Marcell Szasz
- 3 Semmelweis University, Budapest, Hungary.,4 Lund University, Lund, Sweden
| | | | - Hans Bojar
- 6 NextGen Oncology Group, Dusseldorf, Germany
| | | |
Collapse
|
22
|
Riccardi C, Ronchetti S, Nocentini G. Glucocorticoid-induced TNFR-related gene (GITR) as a therapeutic target for immunotherapy. Expert Opin Ther Targets 2018; 22:783-797. [DOI: 10.1080/14728222.2018.1512588] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Carlo Riccardi
- Department of Medicine, University of Perugia, Perugia, Italy
| | | | | |
Collapse
|
23
|
Kopru CZ, Cagnan I, Akar I, Esendagli G, Korkusuz P, Gunel-Ozcan A. Dual Effect of Glucocorticoid-Induced Tumor Necrosis Factor-Related Receptor Ligand Carrying Mesenchymal Stromal Cells on Small Cell Lung Cancer: A Preliminary in vitro Study. Cytotherapy 2018; 20:930-940. [PMID: 30180943 DOI: 10.1016/j.jcyt.2018.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 05/02/2018] [Accepted: 05/02/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND AIMS TNFR family member glucocorticoid-induced tumor necrosis factor-related receptor (GITR/TNFRSF18) activation by its ligand glucocorticoid-induced TNF-related receptor ligand (GITRL) have important roles in proliferation, death and differentiation of cells. Some types of small cell lung cancers (SCLCs) express GITR. Because mesenchymal stromal cells (MSCs) may target tumor cells, we aimed to investigate the effect of MSCs carrying GITRL overexpressing plasmid on the proliferation and viability of a GITR+ SCLC cell line (SCLC-21H) compared with a GITR- SCLC cell line (NCI-H82). METHODS Electroporation was used to transfer pGITRL (GITRL gene carrying plasmid) or pCR3 (mock plasmid) into MSCs. Flow cytometry and semi-quantitative polymerase chain reaction were used to characterize the transfected MSCs. Following SCLC-21H or NCI-H82 cell lines were co-cultured with pGITRL-MSCs. RESULTS Proliferation of NCI-H82 was increased in all types of co-cultures while SCLC-21H cells did not. GITRL expressing MSCs were able to induce cell death of SCLC-21H through the upregulation of SIVA1 apoptosis inducing factor. CONCLUSIONS The influence of MSCs on SCLC cells can vary according to the cancer cell subtypes as obtained in SCLC-21H and NCI-H82 and enabling GITR-GITRL interaction can induce cell death of SCLC cell lines.
Collapse
Affiliation(s)
- Cagla Zubeyde Kopru
- Department of Nanotechnology and Nanomedicine, Graduate School of Science and Engineering, Hacettepe University, Ankara, Turkey
| | - Ilgin Cagnan
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Irem Akar
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Gunes Esendagli
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Aysen Gunel-Ozcan
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
24
|
The promise and challenges of immune agonist antibody development in cancer. Nat Rev Drug Discov 2018; 17:509-527. [PMID: 29904196 DOI: 10.1038/nrd.2018.75] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immune cell functions are regulated by co-inhibitory and co-stimulatory receptors. The first two generations of cancer immunotherapy agents consist primarily of antagonist antibodies that block negative immune checkpoints, such as programmed cell death protein 1 (PD1) and cytotoxic T lymphocyte protein 4 (CTLA4). Looking ahead, there is substantial promise in targeting co-stimulatory receptors with agonist antibodies, and a growing number of these agents are making their way through various stages of development. This Review discusses the key considerations and potential pitfalls of immune agonist antibody design and development, their differentiating features from antagonist antibodies and the landscape of agonist antibodies in clinical development for cancer treatment.
Collapse
|
25
|
Vashist N, Trittel S, Ebensen T, Chambers BJ, Guzmán CA, Riese P. Influenza-Activated ILC1s Contribute to Antiviral Immunity Partially Influenced by Differential GITR Expression. Front Immunol 2018; 9:505. [PMID: 29623077 PMCID: PMC5874297 DOI: 10.3389/fimmu.2018.00505] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 02/26/2018] [Indexed: 01/07/2023] Open
Abstract
Innate lymphoid cells (ILCs) represent diversified subsets of effector cells as well as immune regulators of mucosal immunity and are classified into group 1 ILCs, group 2 ILCs, and group 3 ILCs. Group 1 ILCs encompass natural killer (NK) cells and non-NK ILCs (ILC1s) and mediate their functionality via the rapid production of IFN-γ and TNF-α. The current knowledge of ILC1s mainly associates them to inflammatory processes. Much less is known about their regulation during infection and their capacity to interact with cells of the adaptive immune system. The present study dissected the role of ILC1s during early influenza A virus infection, thereby revealing their impact on the antiviral response. Exploiting in vitro and in vivo H1N1 infection systems, a cross-talk of ILC1s with cells of the innate and the adaptive immunity was demonstrated, which contributes to anti-influenza immunity. A novel association of ILC1 functionality and the expression of the glucocorticoid-induced TNFR-related protein (GITR) was observed, which hints toward a so far undescribed role of GITR in regulating ILC1 responsiveness. Overexpression of GITR inhibits IFN-γ production by ILC1s, whereas partial reduction of GITR expression can reverse this effect, thereby regulating ILC1 functionality. These new insights into ILC1 biology define potential intervention targets to modulate the functional properties of ILC1s, thus contributing toward the development of new immune interventions against influenza.
Collapse
Affiliation(s)
- Neha Vashist
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department of Medicine, Center for Infectious Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Stephanie Trittel
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Benedict J Chambers
- Department of Medicine, Center for Infectious Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Peggy Riese
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
26
|
Pock HL, Wei Bryan TY, Hoon TP, Iqbal J, Ching LY, Tee GY, Wai TK, Chong TH, Wee OK, Wei YK. Standing in the GAP : To formulate a novel radioimmunotherapy regime to improve the long-term outcome in breast cancer. Immunotherapy 2018; 10:255-263. [DOI: 10.2217/imt-2017-0080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The new radioimmunotherapeutic regime GAP15R aims to stimulate glucocorticoid-induced TNF-related protein (G) to overcome Treg suppression; add IFN-α (A) to promote inflammatory milieu; block PD1 (P) to disinhibit T effector cytotoxicity; add IL-15 (15) to enhance danger signals & T-cell expansion; and apply radiation (R) at critical time point to sustain localized inflammation. Patients & methods/materials & methods: This was tested in a murine 4T1 metastatic breast carcinoma model given GAP15R with regular monitoring of tumor volume and complications. Results: We had demonstrated long-term complete remission up to 50% of treated mice, which is not associated with major treatment-related complication, in cases with specific tumor burden. Conclusion: GAP15R is efficacious with potential to be applicable to other tumors.
Collapse
Affiliation(s)
- Ho Liam Pock
- Department of Clinical Pathology, Singapore General Hospital, Singapore
- Department of Hematology, Singapore General Hospital, Singapore
| | - Teo Yi Wei Bryan
- Department of Clinical Research, Singapore General Hospital, Singapore
| | - Tan Puay Hoon
- Division of Pathology, Singapore General Hospital, Singapore
| | - Jabed Iqbal
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Linn Yeh Ching
- Department of Hematology, Singapore General Hospital, Singapore
| | - Goh Yeow Tee
- Department of Hematology, Singapore General Hospital, Singapore
| | - Tan Kar Wai
- Department of Clinical Research, Singapore General Hospital, Singapore
| | - Toh Han Chong
- Department of Medical Oncology, National Cancer Center Singapore, Singapore
| | - Ong Kong Wee
- Department of Surgical Oncology, National Cancer Center Singapore, Singapore
| | - Yeoh Kheng Wei
- Department of Radiation Oncology, National Cancer Center Singapore, Singapore
| |
Collapse
|
27
|
Manson G, Houot R. Next-generation immunotherapies for lymphoma: one foot in the future. Ann Oncol 2018; 29:588-601. [DOI: 10.1093/annonc/mdy032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
28
|
Carter PJ, Lazar GA. Next generation antibody drugs: pursuit of the 'high-hanging fruit'. Nat Rev Drug Discov 2017; 17:197-223. [DOI: 10.1038/nrd.2017.227] [Citation(s) in RCA: 447] [Impact Index Per Article: 55.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
29
|
Morillon YM, Hammond SA, Durham NM, Schlom J, Greiner JW. Enhanced immunotherapy by combining a vaccine with a novel murine GITR ligand fusion protein. Oncotarget 2017; 8:73469-73482. [PMID: 29088720 PMCID: PMC5650275 DOI: 10.18632/oncotarget.20703] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 08/03/2017] [Indexed: 11/25/2022] Open
Abstract
Immunotherapy was significantly enhanced in a murine tumor model by combining a vaccine with a fusion protein designed to target the glucocorticoid-induced tumor necrosis factor (TNF) receptor related gene (GITR) on the surface of T cells. The recombinant poxvirus-based vaccine platform included Modified Vaccinia virus Ankara (rMVA) and fowlpox (rF) vectors as the driver immunogens both engineered to express the human carcinoembryonic antigen (CEA) and three murine costimulatory molecules B7.1, ICAM-1, LFA-3 (designated TRICOM). In previous studies, mice expressing human CEA as a transgene (CEA.Tg mice) vaccinated with rMVA/rF-CEA-TRICOM overcame CEA immune tolerance by inducing anti-CEA‒specific immunity and regression of CEA-expressing tumors. The murine GITR ligand fusion protein (mGITRL-FP) consisted of a mouse IgG2a Fc region, a yeast-derived coiled GCN4 pII and the extracellular GITR-binding domain of murine GITR ligand. The design maximized valency and the potential to agonize the GITR receptor. Combined treatment of the vaccine and mGITRL-FP mediated a more robust tumor regression, leading to sustained improvement in overall survival. The enhanced immunotherapeutic effect was linked to the generation of a strong CD8+ T cell antitumor immune response. A treatment schedule with mGITRL-FP administered prior to the priming rMVA-CEA-TRICOM vaccination was of paramount importance. The mechanism of action for the enhanced antitumor effects resided in the depletion of immune cells, particularly FoxP3+ regulatory T cells, that express high GITR levels following activation. The results provide evidence that targeting GITR with mGITRL-FP in concert with a cancer vaccine represents a potential novel approach to more effective immunotherapy.
Collapse
Affiliation(s)
- Y Maurice Morillon
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - John W Greiner
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
30
|
Cabo M, Offringa R, Zitvogel L, Kroemer G, Muntasell A, Galluzzi L. Trial Watch: Immunostimulatory monoclonal antibodies for oncological indications. Oncoimmunology 2017; 6:e1371896. [PMID: 29209572 PMCID: PMC5706611 DOI: 10.1080/2162402x.2017.1371896] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022] Open
Abstract
The goal of cancer immunotherapy is to establish new or boost pre-existing anticancer immune responses that eradicate malignant cells while generating immunological memory to prevent disease relapse. Over the past few years, immunomodulatory monoclonal antibodies (mAbs) that block co-inhibitory receptors on immune effectors cells - such as cytotoxic T lymphocyte-associated protein 4 (CTLA4), programmed cell death 1 (PDCD1, best known as PD-1) - or their ligands - such as CD274 (best known as PD-L1) - have proven very successful in this sense. As a consequence, many of such immune checkpoint blockers (ICBs) have already entered the clinical practice for various oncological indications. Considerable attention is currently being attracted by a second group of immunomodulatory mAbs, which are conceived to activate co-stimulatory receptors on immune effector cells. Here, we discuss the mechanisms of action of these immunostimulatory mAbs and summarize recent progress in their preclinical and clinical development.
Collapse
Affiliation(s)
- Mariona Cabo
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Rienk Offringa
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
- DKFZ-Bayer Joint Immunotherapeutics Laboratory, German Cancer Research Center, Heidelberg, Germany
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France
- Université Pierre et Marie Curie/Paris VI, Paris
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Aura Muntasell
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
31
|
Kim EJ, Lee JG, Kim JY, Song SH, Joo DJ, Huh KH, Kim MS, Kim BS, Kim YS. Enhanced immune-modulatory effects of thalidomide and dexamethasone co-treatment on T cell subsets. Immunology 2017; 152:628-637. [PMID: 28758197 DOI: 10.1111/imm.12804] [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: 04/02/2017] [Revised: 07/18/2017] [Accepted: 07/24/2017] [Indexed: 12/12/2022] Open
Abstract
Thalidomide (TM) has been reported to have anti-cancer and anti-inflammatory properties, and dexamethasone (DX) is known to reduce inflammation and inhibit production of inflammatory cytokines. Many studies have reported that combinatorial therapy with TM and DX is clinically used to treat multiple myeloma and lupus nephritis, but the mechanism responsible for its effects has not been elucidated. In this study, we determined that TM and DX co-treatment had an enhanced immune-modulatory effect on T cells through regulating the expression of co-stimulatory molecules. Splenic naive T cells from C57BL/6 mice were sort-purified and cultured for CD4+ T cell proliferation and regulatory T (Treg) cell conversion in the presence of TM and/or DX. Following incubation with the drugs, cells were collected and OX40, 4-1BB, and glucocorticoid-induced tumour necrosis factor receptor-related protein (GITR) expression was quantified by flow cytometry. TM (1 or 10 μm) decreased CD4+ T cell proliferation in a dose-dependent manner, whereas TM/DX (0·1 or 1 nm) co-treatment further decreased proliferation. Treg cell populations were preserved following drug treatment. Furthermore, expression of co-stimulatory molecules decreased upon TM/DX co-treatment in effector T (Teff) cells and was preserved in Treg cells. Splenic CD4+ T cells isolated from TM- and DX-treated mice exhibited the same patterns of Teff and Treg cell populations as observed in vitro. Considering the selective effect of TM on different T cell subsets, we suggest that TM may play an immunomodulatory role and that TM/DX combinatorial treatment could further enhance these immunomodulatory effects by regulating GITR, OX40, and 4-1BB expression in CD4+ T cells.
Collapse
Affiliation(s)
- Eun Jee Kim
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Jae Geun Lee
- Department of Transplantation Surgery, Severance Hospital, Yonsei University Health System, Seoul, Korea
| | - Joon Ye Kim
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Hwan Song
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea.,Department of Surgery, College of Medicine, Ewha Women's University, Seoul, Korea
| | - Dong Jin Joo
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea.,Department of Transplantation Surgery, Severance Hospital, Yonsei University Health System, Seoul, Korea
| | - Kyu Ha Huh
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea.,Department of Transplantation Surgery, Severance Hospital, Yonsei University Health System, Seoul, Korea
| | - Myoung Soo Kim
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea.,Department of Transplantation Surgery, Severance Hospital, Yonsei University Health System, Seoul, Korea
| | - Beom Seok Kim
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea.,Department of Internal Medicine (Nephrology), Severance Hospital, Yonsei University Health System, Seoul, Korea
| | - Yu Seun Kim
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea.,Department of Transplantation Surgery, Severance Hospital, Yonsei University Health System, Seoul, Korea
| |
Collapse
|
32
|
Dai J, Fang P, Saredy J, Xi H, Ramon C, Yang W, Choi ET, Ji Y, Mao W, Yang X, Wang H. Metabolism-associated danger signal-induced immune response and reverse immune checkpoint-activated CD40 + monocyte differentiation. J Hematol Oncol 2017; 10:141. [PMID: 28738836 PMCID: PMC5525309 DOI: 10.1186/s13045-017-0504-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/26/2017] [Indexed: 01/16/2023] Open
Abstract
Adaptive immunity is critical for disease progression and modulates T cell (TC) and antigen-presenting cell (APC) functions. Three signals were initially proposed for adaptive immune activation: signal 1 antigen recognition, signal 2 co-stimulation or co-inhibition, and signal 3 cytokine stimulation. In this article, we propose to term signal 2 as an immune checkpoint, which describes interactions of paired molecules leading to stimulation (stimulatory immune checkpoint) or inhibition (inhibitory immune checkpoint) of an immune response. We classify immune checkpoint into two categories: one-way immune checkpoint for forward signaling towards TC only, and two-way immune checkpoint for both forward and reverse signaling towards TC and APC, respectively. Recently, we and others provided evidence suggesting that metabolic risk factors (RF) activate innate and adaptive immunity, involving the induction of immune checkpoint molecules. We summarize these findings and suggest a novel theory, metabolism-associated danger signal (MADS) recognition, by which metabolic RF activate innate and adaptive immunity. We emphasize that MADS activates the reverse immune checkpoint which leads to APC inflammation in innate and adaptive immunity. Our recent evidence is shown that metabolic RF, such as uremic toxin or hyperhomocysteinemia, induced immune checkpoint molecule CD40 expression in monocytes (MC) and elevated serum soluble CD40 ligand (sCD40L) resulting in CD40+ MC differentiation. We propose that CD40+ MC is a novel pro-inflammatory MC subset and a reliable biomarker for chronic kidney disease severity. We summarize that CD40:CD40L immune checkpoint can induce TC and APC activation via forward stimulatory, reverse stimulatory, and TC contact-independent immune checkpoints. Finally, we modeled metabolic RF-induced two-way stimulatory immune checkpoint amplification and discussed potential signaling pathways including AP-1, NF-κB, NFAT, STAT, and DNA methylation and their contribution to systemic and tissue inflammation.
Collapse
Affiliation(s)
- Jin Dai
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian road, Hangzhou, 310006, Zhejiang, China.,Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Pu Fang
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Jason Saredy
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Hang Xi
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Cueto Ramon
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - William Yang
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Eric T Choi
- Department of Surgery, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 210029, China
| | - Wei Mao
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian road, Hangzhou, 310006, Zhejiang, China.
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.,Department of Pharmacology, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA. .,Department of Pharmacology, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
33
|
Sukumar S, Wilson DC, Yu Y, Wong J, Naravula S, Ermakov G, Riener R, Bhagwat B, Necheva AS, Grein J, Churakova T, Mangadu R, Georgiev P, Manfra D, Pinheiro EM, Sriram V, Bailey WJ, Herzyk D, McClanahan TK, Willingham A, Beebe AM, Sadekova S. Characterization of MK-4166, a Clinical Agonistic Antibody That Targets Human GITR and Inhibits the Generation and Suppressive Effects of T Regulatory Cells. Cancer Res 2017; 77:4378-4388. [DOI: 10.1158/0008-5472.can-16-1439] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 11/21/2016] [Accepted: 06/06/2017] [Indexed: 11/16/2022]
|
34
|
Beyond TNF: TNF superfamily cytokines as targets for the treatment of rheumatic diseases. Nat Rev Rheumatol 2017; 13:217-233. [PMID: 28275260 DOI: 10.1038/nrrheum.2017.22] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
TNF blockers are highly efficacious at dampening inflammation and reducing symptoms in rheumatic diseases such as rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis, and also in nonrheumatic syndromes such as inflammatory bowel disease. As TNF belongs to a superfamily of 19 structurally related proteins that have both proinflammatory and anti-inflammatory activity, reagents that disrupt the interaction between proinflammatory TNF family cytokines and their receptors, or agonize the anti-inflammatory receptors, are being considered for the treatment of rheumatic diseases. Biologic agents that block B cell activating factor (BAFF) and receptor activator of nuclear factor-κB ligand (RANKL) have been approved for the treatment of systemic lupus erythematosus and osteoporosis, respectively. In this Review, we focus on additional members of the TNF superfamily that could be relevant for the pathogenesis of rheumatic disease, including those that can strongly promote activity of immune cells or increase activity of tissue cells, as well as those that promote death pathways and might limit inflammation. We examine preclinical mouse and human data linking these molecules to the control of damage in the joints, muscle, bone or other tissues, and discuss their potential as targets for future therapy of rheumatic diseases.
Collapse
|
35
|
Waight JD, Gombos RB, Wilson NS. Harnessing co-stimulatory TNF receptors for cancer immunotherapy: Current approaches and future opportunities. Hum Antibodies 2017; 25:87-109. [PMID: 28085016 DOI: 10.3233/hab-160308] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Co-stimulatory tumor necrosis factor receptors (TNFRs) can sculpt the responsiveness of T cells recognizing tumor-associated antigens. For this reason, agonist antibodies targeting CD137, CD357, CD134 and CD27 have received considerable attention for their therapeutic utility in enhancing anti-tumor immune responses, particularly in combination with other immuno-modulatory antibodies targeting co-inhibitory pathways in T cells. The design of therapeutic antibodies that optimally engage and activate co-stimulatory TNFRs presents an important challenge of how to promote effective anti-tumor immunity while avoiding serious immune-related adverse events. Here we review our current understanding of the expression, signaling and structural features of CD137, CD357, CD134 and CD27, and how this may inform the design of pharmacologically active immuno-modulatory antibodies targeting these receptors. This includes the integration of our emerging knowledge of the role of Fcγ receptors (FcγRs) in facilitating antibody-mediated receptor clustering and forward signaling, as well as promoting immune effector cell-mediated activities. Finally, we bring our current preclinical and clinical knowledge of co-stimulatory TNFR antibodies into the context of opportunities for next generation molecules with improved pharmacologic properties.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/therapeutic use
- Gene Expression Regulation
- Humans
- Immunity, Cellular/drug effects
- Immunotherapy/methods
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/pathology
- Receptors, IgG/agonists
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Receptors, Tumor Necrosis Factor/agonists
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/immunology
- Signal Transduction
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
Collapse
|
36
|
Novel Immunotherapeutic Approaches for Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2016; 8:cancers8100087. [PMID: 27669306 PMCID: PMC5082377 DOI: 10.3390/cancers8100087] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/16/2016] [Accepted: 09/08/2016] [Indexed: 12/11/2022] Open
Abstract
The immune system plays a key role in preventing tumor formation by recognizing and destroying malignant cells. For over a century, researchers have attempted to harness the immune response as a cancer treatment, although this approach has only recently achieved clinical success. Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide and is associated with cigarette smoking, alcohol consumption, betel nut use, and human papillomavirus infection. Unfortunately, worldwide mortality from HNSCC remains high, partially due to limits on therapy secondary to the significant morbidity associated with current treatments. Therefore, immunotherapeutic approaches to HNSCC treatment are attractive for their potential to reduce morbidity while improving survival. However, the application of immunotherapies to this disease has been challenging because HNSCC is profoundly immunosuppressive, resulting in decreased absolute lymphocyte counts, impaired natural killer cell function, reduced antigen-presenting cell function, and a tumor-permissive cytokine profile. Despite these challenges, numerous clinical trials testing the safety and efficacy of immunotherapeutic approaches to HNSCC treatment are currently underway, many of which have produced promising results. This review will summarize immunotherapeutic approaches to HNSCC that are currently undergoing clinical trials.
Collapse
|
37
|
Austin R, Smyth MJ, Lane SW. Harnessing the immune system in acute myeloid leukaemia. Crit Rev Oncol Hematol 2016; 103:62-77. [PMID: 27247119 DOI: 10.1016/j.critrevonc.2016.04.020] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/13/2016] [Accepted: 04/28/2016] [Indexed: 12/13/2022] Open
|
38
|
Kim B, Kim J, Kim E, Lee J, Joo D, Huh K, Kim M, Kim Y. Role of Thalidomide on the Expression of OX40, 4-1BB, and GITR in T Cell Subsets. Transplant Proc 2016; 48:1270-4. [DOI: 10.1016/j.transproceed.2015.12.088] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 12/30/2015] [Indexed: 11/25/2022]
|
39
|
Nocentini G, Cari L, Ronchetti S, Riccardi C. Modulation of tumor immunity: a patent evaluation of WO2015026684A1. Expert Opin Ther Pat 2016; 26:417-25. [DOI: 10.1517/13543776.2016.1118061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
40
|
Redman JM, Hill EM, AlDeghaither D, Weiner LM. Mechanisms of action of therapeutic antibodies for cancer. Mol Immunol 2015; 67:28-45. [PMID: 25911943 PMCID: PMC4529810 DOI: 10.1016/j.molimm.2015.04.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/29/2015] [Accepted: 04/03/2015] [Indexed: 02/06/2023]
Abstract
The therapeutic utility of antibodies and their derivatives is achieved by various means. The FDA has approved several targeted antibodies that disrupt signaling of various growth factor receptors for the treatment of a number of cancers. Rituximab, and other anti-CD20 monoclonal antibodies are active in B cell malignancies. As more experience has been gained with anti-CD20 monoclonal antibodies, the multifactorial nature of their anti-tumor mechanisms has emerged. Other targeted antibodies function to dampen inhibitory checkpoints. These checkpoint inhibitors have recently achieved dramatic results in several cancers, including melanoma. These and related antibodies continue to be investigated in the clinical and pre-clinical settings. Novel antibody structures that target two or more antigens have also made their way into clinical use. Tumor targeted antibodies can also be conjugated to chemo- or radiotherapeutic agents, or catalytic toxins, as a means to deliver toxic payloads to cancer cells. Here we provide a review of these mechanisms and a discussion of their relevance to current and future clinical applications.
Collapse
Affiliation(s)
- J M Redman
- Departments of Oncology and Internal Medicine, Georgetown University Medical Center and Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - E M Hill
- Departments of Oncology and Internal Medicine, Georgetown University Medical Center and Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - D AlDeghaither
- Departments of Oncology and Internal Medicine, Georgetown University Medical Center and Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - L M Weiner
- Departments of Oncology and Internal Medicine, Georgetown University Medical Center and Lombardi Comprehensive Cancer Center, Washington, DC, United States.
| |
Collapse
|
41
|
Kim YH, Shin SM, Choi BK, Oh HS, Kim CH, Lee SJ, Kim KH, Lee DG, Park SH, Kwon BS. Authentic GITR Signaling Fails To Induce Tumor Regression unless Foxp3+ Regulatory T Cells Are Depleted. THE JOURNAL OF IMMUNOLOGY 2015; 195:4721-9. [DOI: 10.4049/jimmunol.1403076] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 09/08/2015] [Indexed: 12/22/2022]
|
42
|
Abstract
Immune responses are tightly regulated via signaling through numerous co-stimulatory and co-inhibitory molecules. Exploitation of these immune checkpoint pathways is one of the mechanisms by which tumors evade and/or escape the immune system. A growing understanding of the biology of immune checkpoints and tumor immunology has led to the development of monoclonal antibodies designed to target co-stimulatory and co-inhibitory molecules in order to re-engage the immune system and restore antitumor immune responses. Anti-cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) antibodies were among the first to be tested in the clinic, and ipilimumab was the first immune checkpoint inhibitor approved for an anticancer indication. Agents targeting the programmed death 1 (PD-1) pathway, either PD-1 or one of its ligands, programmed death ligand 1, are in active clinical development for numerous cancers, including advanced melanoma and lung cancer. Understanding the different mechanisms of action, safety profiles, and response patterns associated with inhibition of the CTLA-4 and PD-1 pathways may improve patient management as these therapies are moved in to the clinical practice setting and may also provide a rationale for combination therapy with different inhibitors. Additional immune checkpoint molecules with therapeutic potential, including lymphocyte activation gene-3 and glucocorticoid-induced tumor necrosis factor receptor-related gene, also have inhibitors in early stages of clinical development. Clinical responses and safety data reported to date on immune checkpoint inhibitors suggest these agents may have the potential to markedly improve outcomes for patients with cancer.
Collapse
Affiliation(s)
- Kent Shih
- Sarah Cannon Research Institute and Tennessee Oncology PLLC, Nashville, TN, 37203, USA,
| | | | | |
Collapse
|
43
|
Yu N, Fu S, Xu Z, Liu Y, Hao J, Zhang A, Wang B. Synergistic antitumor responses by combined GITR activation and sunitinib in metastatic renal cell carcinoma. Int J Cancer 2015; 138:451-62. [PMID: 26239999 DOI: 10.1002/ijc.29713] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 07/09/2015] [Accepted: 07/21/2015] [Indexed: 12/18/2022]
Abstract
Sunitinib, a multitargeted tyrosine kinase inhibitor, is the frontline therapy for renal and gastrointestinal cancers. In view of its well-documented proapoptotic and immunoadjuvant properties, we speculate that combination of Sunitinib and immunotherapy would provide a synergistic antitumor effect. Here, we report that a remarkably synergistic antitumor responses elicited by the combined treatment of Sunitinib and an agonistic antibody against glucocorticoid-induced TNFR related protein (GITR) in a model of metastatic renal cell carcinoma. Sunitinib significantly increased the infiltration, activation, and proliferation and/or cytotoxicity of CD8(+) T cells and NK cells in liver metastatic foci when combined with the anti (α)-GITR agonist, which was associated with treatment-induced prominent upregulation of Th1-biased immune genes in the livers from mice receiving combined therapy versus single treatment. Sunitinib/α-GITR treatment also markedly promoted the maturation, activation and cytokine production of liver-resident macrophages and DCs compared with that achieved by α-GITR or Sunitinib treatment alone in mice. Cell depletion experiments demonstrated that CD8(+) T cells, NK cells and macrophage infiltrating liver metastatic foci all contribute to the antitumor effect induced by combined treatment. Furthermore, mechanistic investigation revealed that Sunitinib treatment reprograms tumor-associated macrophages toward classically activated or "M1" polarization upon GITR stimulation and consequently mounts an antitumor CD8(+) T and NK cell response via inhibiting STAT3 activity. Thus, our findings provide a proof of concept that Sunitinib can synergize with α-GITR treatment to remodel the tumor immune microenvironment to trigger regressions of an established metastatic cancer.
Collapse
Affiliation(s)
- Nengwang Yu
- Department of Urology, General Hospital of Jinan Military Command, Jinan, Shandong, China
| | - Shuai Fu
- Shandong Cancer Hospital & Institute, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Zhonghua Xu
- Department of Urology, Qilu Hospital, Jinan, Shandong, China
| | - Yi Liu
- Department of Urology, General Hospital of Jinan Military Command, Jinan, Shandong, China
| | - Junwen Hao
- Department of Urology, General Hospital of Jinan Military Command, Jinan, Shandong, China
| | - Aimin Zhang
- Department of Urology, General Hospital of Jinan Military Command, Jinan, Shandong, China
| | - Baocheng Wang
- Department of Oncology, General Hospital of Jinan Military Command, Jinan, Shandong, China
| |
Collapse
|
44
|
Pedroza-Gonzalez A, Zhou G, Singh SP, Boor PP, Pan Q, Grunhagen D, de Jonge J, Tran TK, Verhoef C, IJzermans JN, Janssen HLA, Biermann K, Kwekkeboom J, Sprengers D. GITR engagement in combination with CTLA-4 blockade completely abrogates immunosuppression mediated by human liver tumor-derived regulatory T cells ex vivo. Oncoimmunology 2015; 4:e1051297. [PMID: 26587321 DOI: 10.1080/2162402x.2015.1051297] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/01/2015] [Accepted: 05/09/2015] [Indexed: 02/08/2023] Open
Abstract
In liver cancer tumor-infiltrating regulatory T cells (Ti-Treg) are potent suppressors of tumor-specific T-cell responses and express high levels of the Treg-associated molecules cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and glucocorticoid-induced tumor necrosis factor receptor (GITR). In this study, we have evaluated the capacity of GITR-ligation, CTLA-4-blockade and a combination of both treatments to alleviate immunosuppression mediated by Ti-Treg. Using ex vivo isolated cells from individuals with hepatocellular carcinoma (HCC) or liver metastases from colorectal cancer (LM-CRC) we show that treatment with a soluble form of the natural ligand of GITR (GITRL), or with blocking antibodies to CTLA-4, reduces the suppression mediated by human liver tumor-infiltrating CD4+Foxp3+ Treg, thereby restoring proliferation and cytokine production by effector T cells. Importantly, combined treatment with low doses of both molecules exhibited stronger recovery of T cell function compared with either treatment alone. Our data suggest that in patients with primary and secondary liver cancer both GITR-ligation and anti-CTLA-4 mAb can improve the antitumor immunity by abrogating Ti-Treg mediated suppression.
Collapse
Affiliation(s)
- Alexander Pedroza-Gonzalez
- Department of Gastroenterology and Hepatology; Erasmus MC University Medical Center ; Rotterdam, The Netherlands ; Laboratory of Immunology Research, Medicine; Faculty of Higher Studies Iztacala; National Autonomous University of Mexico; FES-Iztacala, UNAM ; Mexico City, Mexico
| | - Guoying Zhou
- Department of Gastroenterology and Hepatology; Erasmus MC University Medical Center ; Rotterdam, The Netherlands
| | - Simar Pal Singh
- Department of Gastroenterology and Hepatology; Erasmus MC University Medical Center ; Rotterdam, The Netherlands
| | - Patrick Pc Boor
- Department of Gastroenterology and Hepatology; Erasmus MC University Medical Center ; Rotterdam, The Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology; Erasmus MC University Medical Center ; Rotterdam, The Netherlands
| | - Dirk Grunhagen
- Department of Surgery; Erasmus MC University Medical Center ; Rotterdam, The Netherlands
| | - Jeroen de Jonge
- Department of Surgery; Erasmus MC University Medical Center ; Rotterdam, The Netherlands
| | - Tc Khe Tran
- Department of Surgery; Erasmus MC University Medical Center ; Rotterdam, The Netherlands
| | - Cornelis Verhoef
- Department of Surgery; Erasmus MC University Medical Center ; Rotterdam, The Netherlands
| | - Jan Nm IJzermans
- Department of Surgery; Erasmus MC University Medical Center ; Rotterdam, The Netherlands
| | - Harry LA Janssen
- Department of Gastroenterology and Hepatology; Erasmus MC University Medical Center ; Rotterdam, The Netherlands
| | - Katharina Biermann
- Department of Pathology; Erasmus MC-University Medical Center ; Rotterdam, The Netherlands
| | - Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology; Erasmus MC University Medical Center ; Rotterdam, The Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology; Erasmus MC University Medical Center ; Rotterdam, The Netherlands
| |
Collapse
|
45
|
Patel MA, Kim JE, Ruzevick J, Lim M. Present and future of immune checkpoint blockade: Monotherapy to adjuvant approaches. World J Immunol 2015; 5:1-15. [DOI: 10.5411/wji.v5.i1.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/23/2014] [Accepted: 11/19/2014] [Indexed: 02/05/2023] Open
Abstract
Immune regulation of aggressive tumor growth is often outpaced by tumor up-regulation of ligands that inhibit effector immune responses through the activation of immune checkpoints. A few of such checkpoints include programmed death-1 (PD-1), cytotoxic T lymphocyte associated antigen-4 (CTLA-4), lymphocyte activation gene-3, T-cell immunoglobulin and mucin protein-3, Glucocorticoid-induced TNFR family-related receptor (GITR), and killer cell immunoglobulin like receptor. With the exception of GITR, after binding to their respective ligands these checkpoints induce down-modulation of immune responses to prevent autoimmunity. However, such immune mechanisms are co-opted by tumors to allow rapid tumor cell proliferation. Pre-clinical studies in antibody blockade of PD-1 and CTLA-4 have led to promising augmentation of effector immune responses in murine tumor models, and human antibodies against PD-1 and CTLA-4 alone or in combination have demonstrated tumor regression in clinical trials. The development of immune checkpoint blockade as a potential future immunotherapy has led to increasing interest in combining treatment modalities. Combination checkpoint blockade with chemotherapy and radiation therapy has shown synergistic effects in pre-clinical and clinical studies, and combination checkpoint blockade with bacterial vaccine vectors have produced increased effector immune responses in pre-clinical models. The future of immune checkpoint blockade may be as a powerful adjuvant alongside the current standard of care.
Collapse
|
46
|
Enhanced CD8 T cell responses through GITR-mediated costimulation resolve chronic viral infection. PLoS Pathog 2015; 11:e1004675. [PMID: 25738498 PMCID: PMC4349659 DOI: 10.1371/journal.ppat.1004675] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 01/09/2015] [Indexed: 01/12/2023] Open
Abstract
Chronic infections are characterized by the inability to eliminate the persisting pathogen and often associated with functional impairment of virus-specific T-cell responses. Costimulation through Glucocorticoid-induced TNFR-related protein (GITR) can increase survival and function of effector T cells. Here, we report that constitutive expression of GITR-ligand (GITRL) confers protection against chronic lymphocytic choriomeningitis virus (LCMV) infection, accelerating recovery without increasing pathology. Rapid viral clearance in GITRL transgenic mice coincided with increased numbers of poly-functional, virus-specific effector CD8+ T cells that expressed more T-bet and reduced levels of the rheostat marker PD-1. GITR triggering also boosted the helper function of virus-specific CD4 T cells already early in the infection, as was evidenced by increased IL-2 and IFNγ production, and more expression of CD40L and T-bet. Importantly, CD4-depletion experiments revealed that the expanded pool of virus-specific effector CD8 T cells and the ensuing viral clearance in LCMV-infected GITRL tg mice was entirely dependent on CD4 T cells. We found no major differences for NK cell and regulatory T cell responses, whereas the humoral response to the virus was increased in GITRL tg mice, but only in the late phase of the infection when the virus was almost eradicated. Based on these findings, we conclude that enhanced GITR-triggering mediates its protective, anti-viral effect on the CD8 T cell compartment by boosting CD4 T cell help. As such, increasing costimulation through GITR may be an attractive strategy to increase anti-viral CTL responses without exacerbating pathology, in particular to persistent viruses such as HIV and HCV. The ability of the immune system to rapidly respond to a viral infection is a prerequisite for the survival of an individual. The immediate reaction of innate immune cells and the subsequent response of antigen-specific lymphocytes is usually effective for rapid neutralization and removal of the invading virus. Yet, such protective immune responses need to be well controlled, as they can cause severe tissue damage that may disable the host more than the infection itself. One way that has evolutionarily been proven effective to deal with this balancing act between protective immunity and prevention of immunopathology is to render virus-specific T cells “exhausted” when the virus cannot be eradicated and the host becomes chronically infected. Exhausted T cells progressively lose their ability to kill other cells and produce different cytokines. The benefit of this exhausted state of anti-viral immunity is that it induces less tissue damage, but the downside is obviously less efficient control over the viral infection. Many immunotherapeutic and vaccination strategies against chronic viral infections are currently dedicated to overcome the exhausted state of the virus-specific T cells and thereby clear the virus. However, the accompanying risk is an exaggerated immune response with overt immunopathology. Here we describe in a mouse model that enhanced triggering through the costimulatory molecule GITR on T cells is able to provide protection upon viral infection and clear an otherwise persistent virus, but importantly without the development of collateral damage due to immunopathology. We show that GITR-mediated costimulation enhances a protective CD8 T cell response, for which CD4 T cell help is required. Our study provides new insights in how a particular costimulatory pathway can be utilized to boost anti-viral immunity, which is highly relevant for the development of safe immunotherapeutic strategies against chronic viral infections in humans.
Collapse
|
47
|
Liu Y, Tang X, Tian J, Zhu C, Peng H, Rui K, Wang Y, Mao C, Ma J, Lu L, Xu H, Wang S. Th17/Treg cells imbalance and GITRL profile in patients with Hashimoto's thyroiditis. Int J Mol Sci 2014; 15:21674-86. [PMID: 25429429 PMCID: PMC4284671 DOI: 10.3390/ijms151221674] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/07/2014] [Accepted: 11/10/2014] [Indexed: 12/31/2022] Open
Abstract
Hashimoto’s thyroiditis (HT) is an organ-specific immune disease characterized by the presence of lymphocytic infiltration and serum autoantibodies. Previous studies have confirmed the critical role of Th17 cells in the pathopoiesis of HT patients. Additionally, regulatory T cells (Treg) display a dysregulatory function in autoimmune disease. The purpose of this study is to investigate the alteration of Th17 and Treg cells in HT patients and explore contributing factors. We found there was an increased ratio of Th17/Treg in HT patients and a positive correlation with autoantibodies (anti-TgAb). In addition, there was an increased level of GITRL, which has been demonstrated to be correlated with the increassement of Th17 cells in the serum and thyroid glands of HT patients; the upregulated serum level of GITRL has a positive correlation with the percentage of Th17 cells in HT patients. In summary, an increase in GITRL may impair the balance of Th17/Treg, and contribute to the pathopoiesis of Hashimoto’s thyroiditis.
Collapse
Affiliation(s)
- Yingzhao Liu
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, China.
| | - Xinyi Tang
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang 212013, China.
| | - Jie Tian
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang 212013, China.
| | - Chenlu Zhu
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang 212013, China.
| | - Huiyong Peng
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, China.
| | - Ke Rui
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang 212013, China.
| | - Yungang Wang
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang 212013, China.
| | - Chaoming Mao
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang 212013, China.
| | - Jie Ma
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang 212013, China.
| | - Liwei Lu
- Department of Pathology and Centre of Infection and Immunology, The University of Hong Kong, Hong Kong 999077, China.
| | - Huaxi Xu
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang 212013, China.
| | - Shengjun Wang
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, China.
| |
Collapse
|
48
|
Petrillo MG, Ronchetti S, Ricci E, Alunno A, Gerli R, Nocentini G, Riccardi C. GITR+ regulatory T cells in the treatment of autoimmune diseases. Autoimmun Rev 2014; 14:117-26. [PMID: 25449679 DOI: 10.1016/j.autrev.2014.10.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 09/28/2014] [Indexed: 01/07/2023]
Abstract
Autoimmune diseases decrease life expectancy and quality of life for millions of women and men. Although treatments can slow disease progression and improve quality of life, all currently available drugs have adverse effects and none of them are curative; therefore, requiring patients to take immunosuppressive drugs for the remainder of their lives. A curative therapy that is safe and effective is urgently needed. We believe that therapies promoting the in vivo expansion of regulatory T cells (Tregs) or injection of in vitro expanded autologous/heterologous Tregs (cellular therapy) can alter the natural history of autoimmune diseases. In this review, we present data from murine and human studies suggesting that 1) glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) plays a crucial role in thymic Treg (tTreg) differentiation and expansion; 2) GITR plays a crucial role in peripheral Treg (pTreg) expansion; 3) in patients with Sjögren syndrome and systemic lupus erythematosus, CD4(+)GITR(+) pTregs are expanded in patients with milder forms of the disease; and 4) GITR is superior to other cell surface markers to differentiate Tregs from other CD4(+) T cells. In this context, we consider two potential new approaches for treating autoimmune diseases consisting of the in vivo expansion of GITR(+) Tregs by GITR-triggering drugs and in vitro expansion of autologous or heterologous GITR(+) Tregs to be infused in patients. Advantages of such an approach, technical problems, and safety issues are discussed.
Collapse
Affiliation(s)
| | - Simona Ronchetti
- Department of Medicine, Section of Pharmacology, University of Perugia, Italy
| | - Erika Ricci
- Department of Medicine, Section of Pharmacology, University of Perugia, Italy
| | - Alessia Alunno
- Department of Medicine, Rheumatology Unit, University of Perugia, Italy
| | - Roberto Gerli
- Department of Medicine, Rheumatology Unit, University of Perugia, Italy
| | - Giuseppe Nocentini
- Department of Medicine, Section of Pharmacology, University of Perugia, Italy.
| | - Carlo Riccardi
- Department of Medicine, Section of Pharmacology, University of Perugia, Italy
| |
Collapse
|
49
|
Nocentini G, Alunno A, Petrillo MG, Bistoni O, Bartoloni E, Caterbi S, Ronchetti S, Migliorati G, Riccardi C, Gerli R. Expansion of regulatory GITR+CD25 low/-CD4+ T cells in systemic lupus erythematosus patients. Arthritis Res Ther 2014; 16:444. [PMID: 25256257 PMCID: PMC4209023 DOI: 10.1186/s13075-014-0444-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 08/28/2014] [Indexed: 12/20/2022] Open
Abstract
Introduction CD4+CD25low/-GITR+ T lymphocytes expressing forkhead box protein P3 (FoxP3) and showing regulatory activity have been recently described in healthy donors. The objective of the study was to evaluate the proportion of CD4+CD25low/-GITR+ T lymphocytes within CD4+ T cells and compare their phenotypic and functional profile with that of CD4+CD25highGITR− T lymphocytes in systemic lupus erythematosus (SLE) patients. Methods The percentage of CD4+CD25low/-GITR+ cells circulating in the peripheral blood (PB) of 32 patients with SLE and 25 healthy controls was evaluated with flow cytometry. CD4+CD25low/-GITR+ cells were isolated with magnetic separation, and their phenotype was compared with that of CD4+CD25highGITR− cells. Regulatory activity of both cell subsets was tested in autologous and heterologous co-cultures after purification through a negative sorting strategy. Results Results indicated that CD4+CD25low/-GITR+ cells are expanded in the PB of 50% of SLE patients. Expansion was observed only in patients with inactive disease. Phenotypic analysis demonstrated that CD4+CD25low/-GITR+ cells display regulatory T-cell (Treg) markers, including FoxP3, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), transforming growth factor-beta (TGF-β), and interleukin (IL)-10. In contrast, CD4+CD25highGITR− cells appear to be activated and express low levels of Treg markers. Functional experiments demonstrated that CD4+CD25low/-GITR+ cells exert a higher inhibitory activity against both autologous and heterologous cells as compared with CD4+CD25highGITR− cells. Suppression is independent of cell contact and is mediated by IL-10 and TGF-β. Conclusions Phenotypic and functional data demonstrate that in SLE patients, CD4+CD25low/-GITR+ cells are fully active Treg cells, possibly representing peripheral Treg (pTreg) that are expanded in patients with inactive disease. These data may suggest a key role of this T-cell subset in the modulation of the abnormal immune response in SLE. Strategies aimed at expanding this Treg subset for therapeutic purpose deserve to be investigated.
Collapse
|
50
|
Sharon E, Polley MY, Bernstein MB, Ahmed M. Immunotherapy and radiation therapy: considerations for successfully combining radiation into the paradigm of immuno-oncology drug development. Radiat Res 2014; 182:252-7. [PMID: 25003314 DOI: 10.1667/rr13707.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
As the immunotherapy of cancer comes of age, adding immunotherapeutic agents to radiation therapy has the potential to improve the outcomes for patients with a wide variety of malignancies. Despite the enormous potential of such combination therapy, laboratory data has been lacking and there is little guidance for pursuing novel treatment strategies. Animal models have significant limitation in combining radiation therapy with immunotherapy and some of the limitations of preclinical models are discussed in this article. In addition to the preclinical challenges, radiation therapy and immunotherapy combinations may have overlapping toxicities, and for both types of therapy, early and late manifestations of toxicity are possible. Given these risks, special attention should be given to the design of the specific Phase I clinical trial that is chosen. In this article, we describe several Phase I design possibilities that may be employed, including the 3 + 3 design (also known as the cohort of 3 design), the continual reassessment method (CRM), and the time-to-event continual reassessment method (TITE-CRM). Efficacy end points for further development of combination therapy must be based on multiple factors, including disease type, stage of disease, the setting of therapy and the goal of therapy. While the designs for future clinical trials will vary, it is clear that these two successful modalities of therapy can and should be combined for the benefit of cancer patients.
Collapse
Affiliation(s)
- Elad Sharon
- a Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | | |
Collapse
|