1
|
Henriksen NL, Jensen PØ, Jensen LK. Immune checkpoint blockade in experimental bacterial infections. J Infect 2025; 90:106391. [PMID: 39756696 DOI: 10.1016/j.jinf.2024.106391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/03/2024] [Accepted: 12/22/2024] [Indexed: 01/07/2025]
Abstract
Immune checkpoint inhibitors designed to reinvigorate immune responses suppressed by cancer cells have revolutionized cancer therapy. Similarities in immune dysregulation between cancer and infectious diseases have prompted investigations into the role of immune checkpoints in infectious diseases, including the therapeutic potential of immune checkpoint blockade and drug repurposing. While most research has centered around viral infections, data for bacterial infections are emerging. This systematic review reports on the in vivo effect of immune checkpoint blockade on bacterial burden and selected immune responses in preclinical studies of bacterial infection, aiming to assess if there could be a rationale for using immunotherapy for bacterial infections. Of the 42 analyzed studies, immune checkpoint blockade reduced the bacterial burden in 60% of studies, had no effect in 28% and increased the bacterial burden in 12%. Findings suggest that the effect of immune checkpoint blockade on bacterial burden is context-dependent and in part relates to the pathogen. Further preclinical research is required to understand how the therapeutic effect of immune checkpoint blockade is mediated in different bacterial infections, and if immune checkpoint blockade can be used as an adjuvant to conventional infection management strategies.
Collapse
Affiliation(s)
- Nicole L Henriksen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark.
| | - Peter Ø Jensen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark; Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Copenhagen, Denmark.
| | - Louise K Jensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
2
|
Eskeland S, Bø-Granquist EG, Stuen S, Lybeck K, Wilhelmsson P, Lindgren PE, Makvandi-Nejad S. Temporal patterns of gene expression in response to inoculation with a virulent Anaplasma phagocytophilum strain in sheep. Sci Rep 2023; 13:20399. [PMID: 37989861 PMCID: PMC10663591 DOI: 10.1038/s41598-023-47801-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/18/2023] [Indexed: 11/23/2023] Open
Abstract
The aim of this study was to characterize the gene expression of host immune- and cellular responses to a Norwegian virulent strain of Anaplasma phagocytophilum, the cause of tick-borne fever in sheep. Ten sheep were intravenously inoculated with a live virulent strain of A. phagocytophilum. Clinical-, observational-, hematological data as well as bacterial load, flow cytometric cell count data from peripheral blood mononuclear cells and host's gene expression post infection was analysed. The transcriptomic data were assessed for pre-set time points over the course of 22 days following the inoculation. Briefly, all inoculated sheep responded with clinical signs of infection 3 days post inoculation and onwards with maximum bacterial load observed on day 6, consistent with tick-borne fever. On days, 3-8, the innate immune responses and effector processes such as IFN1 signaling pathways and cytokine mediated signaling pathways were observed. Several pathways associated with the adaptive immune responses, namely T-cell activation, humoral immune responses, B-cell activation, and T- and B-cell differentiation dominated on the days of 8, 10 and 14. Flow-cytometric analysis of the PBMCs showed a reduction in CD4+CD25+ cells on day 10 and 14 post-inoculation and a skewed CD4:CD8 ratio indicating a reduced activation and proliferation of CD4-T-cells. The genes of important co-stimulatory molecules such as CD28 and CD40LG, important in T- and B-cell activation and proliferation, did not significantly change or experienced downregulation throughout the study. The absence of upregulation of several co-stimulatory molecules might be one possible explanation for the low activation and proliferation of CD4-T-cells during A. phagocytophilum infection, indicating a suboptimal CD4-T-cell response. The upregulation of T-BET, EOMES and IFN-γ on days 8-14 post inoculation, indicates a favoured CD4 Th1- and CD8-response. The dynamics and interaction between CD4+CD25+ and co-stimulatory molecules such as CD28, CD80, CD40 and CD40LG during infection with A. phagocytophilum in sheep needs further investigation in the future.
Collapse
Affiliation(s)
- Sveinung Eskeland
- Department of Production Animal Clinical Science, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Elizabeth Stephansens Vei 15, 1433, Ås, Norway.
| | - Erik G Bø-Granquist
- Department of Production Animal Clinical Science, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Elizabeth Stephansens Vei 15, 1433, Ås, Norway
| | - Snorre Stuen
- Department of Production Animal Clinical Science, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Kyrkjevegen 332/334, 4325, Sandnes, Norway
| | - Kari Lybeck
- Norwegian Veterinary Institute, Elizabeth Stephansens Vei 1, 1433, Ås, Norway
| | - Peter Wilhelmsson
- Division of Clinical Microbiology, Laboratory Medicine, National Reference Laboratory for Borrelia and Other Tick-Borne Bacteria, Region Jönköping County, 553 05, Jönköping, Sweden
| | - Per-Eric Lindgren
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, 581 83, Linköping, Sweden
| | | |
Collapse
|
3
|
Nandi D, Pathak S, Verma T, Singh M, Chattopadhyay A, Thakur S, Raghavan A, Gokhroo A, Vijayamahantesh. T cell costimulation, checkpoint inhibitors and anti-tumor therapy. J Biosci 2021. [PMID: 32345776 DOI: 10.1007/s12038-020-0020-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The hallmarks of the adaptive immune response are specificity and memory. The cellular response is mediated by T cells which express cell surface T cell receptors (TCRs) that recognize peptide antigens in complex with major histocompatibility complex (MHC) molecules on antigen presenting cells (APCs). However, binding of cognate TCRs with MHC-peptide complexes alone (signal 1) does not trigger optimal T cell activation. In addition to signal 1, the binding of positive and negative costimulatory receptors to their ligands modulates T cell activation. This complex signaling network prevents aberrant activation of T cells. CD28 is the main positive costimulatory receptor on naı¨ve T cells; upon activation, CTLA4 is induced but reduces T cell activation. Further studies led to the identification of additional negative costimulatory receptors known as checkpoints, e.g. PD1. This review chronicles the basic studies in T cell costimulation that led to the discovery of checkpoint inhibitors, i.e. antibodies to negative costimulatory receptors (e.g. CTLA4 and PD1) which reduce tumor growth. This discovery has been recognized with the award of the 2018 Nobel prize in Physiology/Medicine. This review highlights the structural and functional roles of costimulatory receptors, the mechanisms by which checkpoint inhibitors work, the challenges encountered and future prospects.
Collapse
Affiliation(s)
- Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bengaluru 560 012, India
| | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Cytotoxic T-Lymphocyte-Associated Antigen 4 (CTLA-4)- and Programmed Death 1 (PD-1)-Mediated Regulation of Monofunctional and Dual Functional CD4 + and CD8 + T-Cell Responses in a Chronic Helminth Infection. Infect Immun 2019; 87:IAI.00469-19. [PMID: 31570560 DOI: 10.1128/iai.00469-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/23/2019] [Indexed: 11/20/2022] Open
Abstract
Chronic helminth infections are known to be associated with the modulation of antigen-specific T-cell responses. Strongyloides stercoralis infection is characterized by the downmodulation of antigen-specific Th1 and Th17 responses and the upregulation of Th2 and Th9 responses. Immune homeostasis is partially maintained by negative regulators of T-cell activation, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death 1 (PD-1), which dampen effector responses during chronic infections. However, their roles in S. stercoralis infection are yet to be defined. Therefore, we sought to determine the role of CTLA-4 and PD-1 in regulating CD4+ and CD8+ T-cell responses and examined the frequencies of monofunctional and dual functional Th1/T cytotoxic type 1 (Tc1), Th17/Tc17, Th2/Tc2, and Th9/Tc9 cells in S. stercoralis infection in 15 infected individuals stimulated with parasite antigen following CTLA-4 or PD-1 blockade. Our data reveal that CTLA-4 or PD-1 blockade results in significantly enhanced frequencies of monofunctional and dual functional Th1/Tc1 and Th17/Tc17 cells and, in contrast, diminishes the frequencies of monofunctional and dual functional Th2/Tc2 and Th9/Tc9 cells with parasite antigen stimulation in whole-blood cultures. Thus, we demonstrate that CTLA-4 and PD-1 limit the induction of particular T-cell subsets in S. stercoralis infection, which suggests the importance of CTLA-4 and PD-1 in immune modulation in a chronic helminth infection.
Collapse
|
5
|
Tang CL, Pan Q, Xie YP, Xiong Y, Zhang RH, Huang J. Effect of Cytotoxic T-Lymphocyte Antigen-4 on the Efficacy of the Fatty Acid-Binding Protein Vaccine Against Schistosoma japonicum. Front Immunol 2019; 10:1022. [PMID: 31134084 PMCID: PMC6514142 DOI: 10.3389/fimmu.2019.01022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/23/2019] [Indexed: 01/13/2023] Open
Abstract
The present study evaluated the impact of blocking cytotoxic T-lymphocyte antigen-4 (CTLA-4) activity on the protective effect elicited by the fatty acid binding protein (FABP) vaccine against Schistosoma japonicum infection. Mice were randomly divided into uninfected, infected control, anti-CTLA-4 monoclonal antibody (anti-CTLA-4 mAb), FABP, and combination (anti-CTLA-4 mAb and FABP) groups. An assessment of the S. japonicum worm and egg burden in the infected mice revealed that the worm reduction-rate induced by FABP administration was increased from 26.58 to 54.61% by co-administration of the monoclonal anti-CTLA antibody (anti-CTLA-4 mAb). Furthermore, the regulatory T cell (Treg) percentage was significantly increased in mice after administration of the anti-CTLA-4 mAb, but not the FABP vaccine, and elevated levels of the cytokines interferon (IFN)-γ, interleukin (IL)-2, IL-4, and IL-5 were observed in infected mice that were administered the anti-CTLA-4 mAb. Notably, the diameter of egg granulomas in the anti-CTLA-4 mAb and combination groups was significantly increased compared to that observed in the infected control group. Together, these results suggest that co-administering the FABP vaccine and anti-CTLA-4 treatment may have synergistically increased the immunoprotective effect of the FABP vaccine by promoting T-helper 1-type immune responses, while incurring increased tissue damage.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Qun Pan
- Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Ya-Ping Xie
- Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Ying Xiong
- Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Rong-Hui Zhang
- Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Jin Huang
- Department of Clinical Laboratory, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Poirier N, Chevalier M, Mary C, Hervouet J, Minault D, Baker P, Ville S, Le Bas-Bernardet S, Dilek N, Belarif L, Cassagnau E, Scobie L, Blancho G, Vanhove B. Selective CD28 Antagonist Blunts Memory Immune Responses and Promotes Long-Term Control of Skin Inflammation in Nonhuman Primates. THE JOURNAL OF IMMUNOLOGY 2015; 196:274-83. [PMID: 26597009 DOI: 10.4049/jimmunol.1501810] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 10/28/2015] [Indexed: 11/19/2022]
Abstract
Novel therapies that specifically target activation and expansion of pathogenic immune cell subsets responsible for autoimmune attacks are needed to confer long-term remission. Pathogenic cells in autoimmunity include memory T lymphocytes that are long-lived and present rapid recall effector functions with reduced activation requirements. Whereas the CD28 costimulation pathway predominantly controls priming of naive T cells and hence generation of adaptive memory cells, the roles of CD28 costimulation on established memory T lymphocytes and the recall of memory responses remain controversial. In contrast to CD80/86 antagonists (CTLA4-Ig), selective CD28 antagonists blunt T cell costimulation while sparing CTLA-4 and PD-L1-dependent coinhibitory signals. Using a new selective CD28 antagonist, we showed that Ag-specific reactivation of human memory T lymphocytes was prevented. Selective CD28 blockade controlled both cellular and humoral memory recall in nonhuman primates and induced long-term Ag-specific unresponsiveness in a memory T cell-mediated inflammatory skin model. No modification of memory T lymphocytes subsets or numbers was observed in the periphery, and importantly no significant reactivation of quiescent viruses was noticed. These findings indicate that pathogenic memory T cell responses are controlled by both CD28 and CTLA-4/PD-L1 cosignals in vivo and that selectively targeting CD28 would help to promote remission of autoimmune diseases and control chronic inflammation.
Collapse
Affiliation(s)
- Nicolas Poirier
- INSERM UMR 1064, Institut de Transplantation Urologie Néphrologie, Université de Nantes, 44000 Nantes, France; Effimune, 44000 Nantes, France
| | - Melanie Chevalier
- INSERM UMR 1064, Institut de Transplantation Urologie Néphrologie, Université de Nantes, 44000 Nantes, France; Centre Hospitalier Universitaire, 44000 Nantes, France; and
| | - Caroline Mary
- INSERM UMR 1064, Institut de Transplantation Urologie Néphrologie, Université de Nantes, 44000 Nantes, France; Effimune, 44000 Nantes, France
| | - Jeremy Hervouet
- INSERM UMR 1064, Institut de Transplantation Urologie Néphrologie, Université de Nantes, 44000 Nantes, France
| | - David Minault
- INSERM UMR 1064, Institut de Transplantation Urologie Néphrologie, Université de Nantes, 44000 Nantes, France
| | - Paul Baker
- Department of Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, United Kingdom
| | - Simon Ville
- INSERM UMR 1064, Institut de Transplantation Urologie Néphrologie, Université de Nantes, 44000 Nantes, France
| | - Stephanie Le Bas-Bernardet
- INSERM UMR 1064, Institut de Transplantation Urologie Néphrologie, Université de Nantes, 44000 Nantes, France; Centre Hospitalier Universitaire, 44000 Nantes, France; and
| | - Nahzli Dilek
- INSERM UMR 1064, Institut de Transplantation Urologie Néphrologie, Université de Nantes, 44000 Nantes, France; Effimune, 44000 Nantes, France
| | - Lyssia Belarif
- INSERM UMR 1064, Institut de Transplantation Urologie Néphrologie, Université de Nantes, 44000 Nantes, France; Effimune, 44000 Nantes, France
| | | | - Linda Scobie
- Department of Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, United Kingdom
| | - Gilles Blancho
- INSERM UMR 1064, Institut de Transplantation Urologie Néphrologie, Université de Nantes, 44000 Nantes, France; Centre Hospitalier Universitaire, 44000 Nantes, France; and
| | - Bernard Vanhove
- INSERM UMR 1064, Institut de Transplantation Urologie Néphrologie, Université de Nantes, 44000 Nantes, France; Effimune, 44000 Nantes, France;
| |
Collapse
|
7
|
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
|
8
|
Pick J, Arra A, Lingel H, Hegel JK, Huber M, Nishanth G, Jorch G, Fischer KD, Schlüter D, Tedford K, Brunner-Weinzierl MC. CTLA-4 (CD152) enhances the Tc17 differentiation program. Eur J Immunol 2014; 44:2139-52. [PMID: 24723371 DOI: 10.1002/eji.201343497] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 02/21/2014] [Accepted: 03/28/2014] [Indexed: 01/24/2023]
Abstract
Although CD8(+) T cells that produce IL-17 (Tc17 cells) have been linked to host defense, Tc17 cells show reduced cytotoxic activity, which is the characteristic function of CD8(+) T cells. Here, we show that CTLA-4 enhances the frequency of IL-17 in CD8(+) T cells, indicating that CTLA-4 (CD152) specifically promotes Tc17 differentiation. Simultaneous stimulation of CTLA-4(+/+) and CTLA-4(-/-) T cells in cocultures and agonistic CTLA-4 stimulation unambiguously revealed a cell-intrinsic mechanism for IL-17 control by CTLA-4. The quality of CTLA-4-induced Tc17 cells was tested in vivo, utilizing infection with the facultative intracellular bacterium Listeria monocytogenes (LM). Unlike CTLA-4(+/+) Tc17 cells, CTLA-4(-/-) were nearly as efficient as Tc1 CTLA-4(+/+) cells in LM clearance. Additionally, adoptively transferred CTLA-4(-/-) Tc17 cells expressed granzyme B after rechallenge, and produced Tc1 cytokines such as IFN-γ and TNF-α, which strongly correlate with bacterial clearance. CTLA-4(+/+) Tc17 cells demonstrated a high-quality Tc17 differentiation program ex vivo, which was also evident in isolated IL-17-secreting Tc17 cells, with CTLA-4-mediated enhanced upregulation of Tc17-related molecules such as IL-17A, RORγt, and IRF-4. Our results show that CTLA-4 promotes Tc17 differentiation that results in robust Tc17 responses. Its inactivation might therefore represent a central therapeutic target to enhance clearance of infection.
Collapse
Affiliation(s)
- Jonas Pick
- Department of Pediatrics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
McBerry C, Dias A, Shryock N, Lampe K, Gutierrez FRS, Boon L, De'Broski RH, Aliberti J. PD-1 modulates steady-state and infection-induced IL-10 production in vivo. Eur J Immunol 2013; 44:469-79. [PMID: 24165808 PMCID: PMC3955717 DOI: 10.1002/eji.201343658] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 09/17/2013] [Accepted: 10/22/2013] [Indexed: 12/31/2022]
Abstract
Programmed death-1 (PD-1) plays an important role in mediating immune tolerance through mechanisms that remain unclear. Herein, we investigated whether PD-1 prevents excessive host tissue damage during infection with the protozoan parasite, Toxoplasma gondii. Surprisingly, our results demonstrate that PD-1-deficient mice have increased susceptibility to T. gondii, with increased parasite cyst counts along with reduced type-1 cytokine responses (IL-12 and IFN-γ). PD-1⁻/⁻ DCs showed no cell intrinsic defect in IL-12 production in vitro. Instead, PD-1 neutralization via genetic or pharmacological approaches resulted in a striking increase in IL-10 release, which impaired type-1-inflammation during infection. Our results indicate that the absence of PD-1 increases IL-10 production even in the absence of infection. Although the possibility that such increased IL-10 protects against autoimmune damage is speculative, our results show that IL-10 suppresses the development of protective Th1 immune response after T. gondii infection.
Collapse
Affiliation(s)
- Cortez McBerry
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Hafalla JCR, Claser C, Couper KN, Grau GE, Renia L, de Souza JB, Riley EM. The CTLA-4 and PD-1/PD-L1 inhibitory pathways independently regulate host resistance to Plasmodium-induced acute immune pathology. PLoS Pathog 2012; 8:e1002504. [PMID: 22319445 PMCID: PMC3271068 DOI: 10.1371/journal.ppat.1002504] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 12/11/2011] [Indexed: 12/20/2022] Open
Abstract
The balance between pro-inflammatory and regulatory immune responses in determining optimal T cell activation is vital for the successful resolution of microbial infections. This balance is maintained in part by the negative regulators of T cell activation, CTLA-4 and PD-1/PD-L, which dampen effector responses during chronic infections. However, their role in acute infections, such as malaria, remains less clear. In this study, we determined the contribution of CTLA-4 and PD-1/PD-L to the regulation of T cell responses during Plasmodium berghei ANKA (PbA)-induced experimental cerebral malaria (ECM) in susceptible (C57BL/6) and resistant (BALB/c) mice. We found that the expression of CTLA-4 and PD-1 on T cells correlates with the extent of pro-inflammatory responses induced during PbA infection, being higher in C57BL/6 than in BALB/c mice. Thus, ECM develops despite high levels of expression of these inhibitory receptors. However, antibody-mediated blockade of either the CTLA-4 or PD-1/PD-L1, but not the PD-1/PD-L2, pathways during PbA-infection in ECM-resistant BALB/c mice resulted in higher levels of T cell activation, enhanced IFN-γ production, increased intravascular arrest of both parasitised erythrocytes and CD8+ T cells to the brain, and augmented incidence of ECM. Thus, in ECM-resistant BALB/c mice, CTLA-4 and PD-1/PD-L1 represent essential, independent and non-redundant pathways for maintaining T cell homeostasis during a virulent malaria infection. Moreover, neutralisation of IFN-γ or depletion of CD8+ T cells during PbA infection was shown to reverse the pathologic effects of regulatory pathway blockade, highlighting that the aetiology of ECM in the BALB/c mice is similar to that in C57BL/6 mice. In summary, our results underscore the differential and complex regulation that governs immune responses to malaria parasites. T cells are part of the body's defense system in response to infection. However, once the infection has been suitably controlled, these T cells must be switched off. Inhibitory pathways, such as CTLA-4 and PD-1, are known to send the ‘turn off’ signal to T cells during chronic infections. However, their roles in acute infections, such as malaria, are unclear. We compared the function of these inhibitory pathways in mice that are either susceptible or resistant to severe malarial disease (cerebral malaria). Strikingly, we found that receptors for CTLA-4 and PD-1 are more highly expressed in T cells from susceptible mice than from resistant mice. Therefore, cerebral malaria develops despite the high expression of these inhibitory receptors. Moreover, we demonstrated that blocking these inhibitory receptors in the resistant mice increased the function of T cells, which in turn led to the characteristic signs of cerebral malaria. Finally, reminiscent of what is known for the susceptible strain, we confirmed that certain T cells (CD8+) and molecules (IFN-γ) are crucial to the development of cerebral malaria in the otherwise resistant mice. Thus, the CTLA-4 and PD-1 inhibitory pathways have essential, independent and non-redundant roles in regulating the body's complex response to malaria.
Collapse
Affiliation(s)
- Julius Clemence R Hafalla
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
11
|
Olino K, Wada S, Edil BH, Pan X, Meckel K, Weber W, Slansky J, Tamada K, Lauer P, Brockstedt D, Pardoll D, Schulick R, Yoshimura K. Tumor-associated antigen expressing Listeria monocytogenes induces effective primary and memory T-cell responses against hepatic colorectal cancer metastases. Ann Surg Oncol 2011; 19 Suppl 3:S597-607. [PMID: 21979110 PMCID: PMC4498288 DOI: 10.1245/s10434-011-2037-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Indexed: 12/11/2022]
Abstract
Purpose Despite advances in therapy for the treatment of metastatic colorectal cancer, many patients die of hepatic disease. Current immunotherapeutic strategies are likely limited by inhibitory signals from the tumor. To successfully eliminate tumor deposits within an organ, an appropriate immunologic milieu to amplify antitumor responses must be developed. Methods We used a murine model utilizing the CT26 colon cancer cell line to analyze primary and memory tumor-specific T-cell responses induced by an attenuated actin A and internalin B deleted immunodominant tumor-associated antigen expressing strain of Listeria monocytogenes for the treatment of metastatic colorectal cancer. Results Treatment of mice bearing established hepatic metastases with this L. monocytogenes strain led to the generation of a strong initial tumor-specific cytotoxic CD8+ T-cell response that successfully treated 90% of animals. Tumor antigen-specific central and effector memory T cells were also generated and protected against tumor rechallenge. These cell populations, when measured before and after tumor rechallenge, showed a marked expansion of antigen-specific effector CD8+ effector memory T cells. This strain of L. monocytogenes was able to down-modulate the expression of the immune checkpoint molecule, PD-1, within the tumor microenvironment but had variable effects on CTLA-4 expression. Conclusions This L. monocytogenes strain generated a highly effective antitumor T-cell response, providing a basis for the development of this vaccine platform in patients with liver metastases. Electronic supplementary material The online version of this article (doi:10.1245/s10434-011-2037-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kelly Olino
- Department of Surgery, Sidney Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Johanns TM, Ertelt JM, Rowe JH, Way SS. Regulatory T cell suppressive potency dictates the balance between bacterial proliferation and clearance during persistent Salmonella infection. PLoS Pathog 2010; 6:e1001043. [PMID: 20714351 PMCID: PMC2920851 DOI: 10.1371/journal.ppat.1001043] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2010] [Accepted: 07/13/2010] [Indexed: 12/31/2022] Open
Abstract
The pathogenesis of persistent infection is dictated by the balance between opposing immune activation and suppression signals. Herein, virulent Salmonella was used to explore the role and potential importance of Foxp3-expressing regulatory T cells in dictating the natural progression of persistent bacterial infection. Two distinct phases of persistent Salmonella infection are identified. In the first 3-4 weeks after infection, progressively increasing bacterial burden was associated with delayed effector T cell activation. Reciprocally, at later time points after infection, reductions in bacterial burden were associated with robust effector T cell activation. Using Foxp3(GFP) reporter mice for ex vivo isolation of regulatory T cells, we demonstrate that the dichotomy in infection tempo between early and late time points is directly paralleled by drastic changes in Foxp3(+) Treg suppressive potency. In complementary experiments using Foxp3(DTR) mice, the significance of these shifts in Treg suppressive potency on infection outcome was verified by enumerating the relative impacts of regulatory T cell ablation on bacterial burden and effector T cell activation at early and late time points during persistent Salmonella infection. Moreover, Treg expression of CTLA-4 directly paralleled changes in suppressive potency, and the relative effects of Treg ablation could be largely recapitulated by CTLA-4 in vivo blockade. Together, these results demonstrate that dynamic regulation of Treg suppressive potency dictates the course of persistent bacterial infection.
Collapse
Affiliation(s)
- Tanner M. Johanns
- Departments of Pediatrics and Microbiology, University of Minnesota School of Medicine, Center for Microbiology and Infectious Disease Translational Research, Minneapolis, Minnesota, United States of America
| | - James M. Ertelt
- Departments of Pediatrics and Microbiology, University of Minnesota School of Medicine, Center for Microbiology and Infectious Disease Translational Research, Minneapolis, Minnesota, United States of America
| | - Jared H. Rowe
- Departments of Pediatrics and Microbiology, University of Minnesota School of Medicine, Center for Microbiology and Infectious Disease Translational Research, Minneapolis, Minnesota, United States of America
| | - Sing Sing Way
- Departments of Pediatrics and Microbiology, University of Minnesota School of Medicine, Center for Microbiology and Infectious Disease Translational Research, Minneapolis, Minnesota, United States of America
| |
Collapse
|
13
|
Chi N, Maranchie JK, Appleman LJ, Storkus WJ. Update on vaccine development for renal cell cancer. Res Rep Urol 2010; 2:125-41. [PMID: 24198621 PMCID: PMC3703676 DOI: 10.2147/rru.s7242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) remains a significant health concern that frequently presents as metastatic disease at the time of initial diagnosis. Current first-line therapeutics for the advanced-stage RCC include antiangiogenic drugs that have yielded high rates of objective clinical response; however, these tend to be transient in nature, with many patients becoming refractory to chronic treatment with these agents. Adjuvant immunotherapies remain viable candidates to sustain disease-free and overall patient survival. In particular, vaccines designed to optimize the activation, maintenance, and recruitment of specific immunity within or into the tumor site continue to evolve. Based on the integration of increasingly refined immunomonitoring systems in both translational models and clinical trials, allowing for the improved understanding of treatment mechanism(s) of action, further refined (combinational) vaccine protocols are currently being developed and evaluated. This review provides a brief history of RCC vaccine development, discusses the successes and limitations in such approaches, and provides a rationale for developing combinational vaccine approaches that may provide improved clinical benefits to patients with RCC.
Collapse
Affiliation(s)
- Nina Chi
- Department of immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | | | | | | |
Collapse
|