1
|
Abstract
Natural killer T (NKT) cells are a population of innate-like T cells capable of enhancing both innate and adaptive immune responses. Co-delivering an NKT cell agonist and antigen can provide molecular signals to antigen-presenting cells, such as dendritic and B cells, that facilitate strong antigen-specific adaptive immune responses. Accordingly, there has been a significant number of developmental NKT cell-dependent vaccine therapies developed, particularly in the last decade, with many incorporating cancer antigens. In this review, we summarize studies that chemically conjugate the NKT cell agonist and antigen as an effective strategy for agonist-antigen co-delivery to drive antitumor responses.
Collapse
Affiliation(s)
- Benjamin J Compton
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt 5010, New Zealand
| | - Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt 5010, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1010, New Zealand
| |
Collapse
|
2
|
Gu X, Chu Q, Ma X, Wang J, Chen C, Guan J, Ren Y, Wu S, Zhu H. New insights into iNKT cells and their roles in liver diseases. Front Immunol 2022; 13:1035950. [PMID: 36389715 PMCID: PMC9643775 DOI: 10.3389/fimmu.2022.1035950] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/14/2022] [Indexed: 08/29/2023] Open
Abstract
Natural killer T cells (NKTs) are an important part of the immune system. Since their discovery in the 1990s, researchers have gained deeper insights into the physiology and functions of these cells in many liver diseases. NKT cells are divided into two subsets, type I and type II. Type I NKT cells are also named iNKT cells as they express a semi-invariant T cell-receptor (TCR) α chain. As part of the innate immune system, hepatic iNKT cells interact with hepatocytes, macrophages (Kupffer cells), T cells, and dendritic cells through direct cell-to-cell contact and cytokine secretion, bridging the innate and adaptive immune systems. A better understanding of hepatic iNKT cells is necessary for finding new methods of treating liver disease including autoimmune liver diseases, alcoholic liver diseases (ALDs), non-alcoholic fatty liver diseases (NAFLDs), and liver tumors. Here we summarize how iNKT cells are activated, how they interact with other cells, and how they function in the presence of liver disease.
Collapse
Affiliation(s)
- Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Ma
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jing Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Guan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanli Ren
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shanshan Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haihong Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Li YQ, Yan C, Wang XF, Xian MY, Zou GQ, Gao XF, Luo R, Liu Z. A New iNKT-Cell Agonist-Adjuvanted SARS-CoV-2 Subunit Vaccine Elicits Robust Neutralizing Antibody Responses. ACS Infect Dis 2022; 8:2161-2170. [PMID: 36043698 DOI: 10.1021/acsinfecdis.2c00296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Adjuvants are essential components of vaccines. Invariant natural killer T (iNKT) cells are a distinct subset of T cells that function to bridge the innate and adaptive immunities and are capable of mediating strong and rapid responses to a range of diseases, including cancer and infectious disease. An increasing amount of evidence suggests that iNKT cells can help fight viral infection. In particular, iNKT-secreting IL-4 is a key mediator of humoral immunity and has a positive correlation with the levels of neutralizing antibodies. As iNKT cell agonists, αGC glycolipid (α-galactosylceramide, or KRN7000) and its analogues as vaccine adjuvants have begun to provide vaccinologists with a new toolset. Herein we found that a new iNKT-cell agonist αGC-CPOEt elicited a strong cytokine response with increased IL-4 production. Remarkably, after three immunizations, SARS-CoV-2 RBD-Fc adjuvanted by αGC-CPOEt evoked robust neutralizing antibody responses that were about 5.5-fold more than those induced by αGC/RBD-Fc and 25-fold greater than those induced by unadjuvanted RBD-Fc. These findings imply that αGC-CPOEt could be investigated further as a new COVID-19 vaccine adjuvant to prevent current and future infectious disease outbreaks.
Collapse
Affiliation(s)
- Ya-Qian Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Cheng Yan
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Xi-Feng Wang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Mao-Ying Xian
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Guo-Qing Zou
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Xiao-Fei Gao
- Jiangxi Key Laboratory for Mass Spectrometry and Instrumentation, East China University of Technology, Nanchang, Jiangxi 330013, PR China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Zheng Liu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| |
Collapse
|
4
|
Burn OK, Farrand K, Pritchard T, Draper S, Tang CW, Mooney AH, Schmidt AJ, Yang SH, Williams GM, Brimble MA, Kandasamy M, Marshall AJ, Clarke K, Painter GF, Hermans IF, Weinkove R. Glycolipid-peptide conjugate vaccines elicit CD8 + T-cell responses and prevent breast cancer metastasis. Clin Transl Immunology 2022; 11:e1401. [PMID: 35795321 PMCID: PMC9250805 DOI: 10.1002/cti2.1401] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 11/08/2022] Open
Abstract
Objectives Metastasis is the principal cause of breast cancer mortality. Vaccines targeting breast cancer antigens have yet to demonstrate clinical efficacy, and there remains an unmet need for safe and effective treatment to reduce the risk of metastasis, particularly for people with triple-negative breast cancer (TNBC). Certain glycolipids can act as vaccine adjuvants by specifically stimulating natural killer T (NKT) cells to provide a universal form of T-cell help. Methods We designed and made a series of conjugate vaccines comprising a prodrug of the NKT cell-activating glycolipid α-galactosylceramide covalently linked to tumor-expressed peptides, and assessed these using E0771- and 4T1-based breast cancer models in vivo. We employed peptides from the model antigen ovalbumin and from clinically relevant breast cancer antigens HER2 and NY-ESO-1. Results Glycolipid-peptide conjugate vaccines that activate NKT cells led to antigen-presenting cell activation, induced inflammatory cytokines, and, compared with peptide alone or admixed peptide and α-galactosylceramide, specifically enhanced CD8+ T-cell responses against tumor-associated peptides. Primary tumor growth was delayed by vaccination in all tumor models. Using 4T1-based cell lines expressing HER2 or NY-ESO-1, a single administration of the relevant conjugate vaccine prevented tumor colonisation of the lung following intravenous inoculation of tumor cells or spontaneous metastasis from breast, respectively. Conclusion Glycolipid-peptide conjugate vaccines that activate NKT cells prevent lung metastasis in breast cancer models and warrant investigation as adjuvant therapies for high-risk breast cancer.
Collapse
Affiliation(s)
- Olivia K Burn
- Malaghan Institute of Medical Research Wellington New Zealand.,Department of Pathology & Molecular Medicine University of Otago Wellington Wellington New Zealand
| | - Kathryn Farrand
- Malaghan Institute of Medical Research Wellington New Zealand
| | - Tara Pritchard
- Malaghan Institute of Medical Research Wellington New Zealand
| | - Sarah Draper
- Ferrier Research Institute Victoria University of Wellington Wellington New Zealand
| | - Ching-Wen Tang
- Malaghan Institute of Medical Research Wellington New Zealand
| | - Anna H Mooney
- Malaghan Institute of Medical Research Wellington New Zealand
| | | | - Sung H Yang
- School of Chemical Sciences University of Auckland Auckland New Zealand
| | | | - Margaret A Brimble
- School of Chemical Sciences University of Auckland Auckland New Zealand.,School of Biological Sciences University of Auckland Auckland New Zealand.,Maurice Wilkins Centre Auckland New Zealand
| | - Matheswaran Kandasamy
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine University of Oxford Oxford UK
| | - Andrew J Marshall
- Ferrier Research Institute Victoria University of Wellington Wellington New Zealand
| | - Kate Clarke
- Wellington Blood & Cancer Centre Capital & Coast District Health Board Wellington New Zealand
| | - Gavin F Painter
- Ferrier Research Institute Victoria University of Wellington Wellington New Zealand.,Maurice Wilkins Centre Auckland New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research Wellington New Zealand.,Maurice Wilkins Centre Auckland New Zealand
| | - Robert Weinkove
- Malaghan Institute of Medical Research Wellington New Zealand.,Department of Pathology & Molecular Medicine University of Otago Wellington Wellington New Zealand.,Wellington Blood & Cancer Centre Capital & Coast District Health Board Wellington New Zealand
| |
Collapse
|
5
|
Prasit KK, Ferrer-Font L, Burn OK, Anderson RJ, Compton BJ, Schmidt AJ, Mayer JU, Chen CJJ, Dasyam N, Ritchie DS, Godfrey DI, Mattarollo SR, Dundar PR, Painter GF, Hermans IF. Intratumoural administration of an NKT cell agonist with CpG promotes NKT cell infiltration associated with an enhanced antitumour response and abscopal effect. Oncoimmunology 2022; 11:2081009. [PMID: 35712122 PMCID: PMC9196710 DOI: 10.1080/2162402x.2022.2081009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intratumoural administration of unmethylated cytosine-phosphate-guanine motifs (CpG) to stimulate toll-like receptor (TLR)-9 has been shown to induce tumour regression in preclinical studies and some efficacy in the clinic. Because activated natural killer T (NKT) cells can cooperate with pattern-recognition via TLRs to improve adaptive immune responses, we assessed the impact of combining a repeated dosing regimen of intratumoural CpG with a single intratumoural dose of the NKT cell agonist α-galactosylceramide (α-GalCer). The combination was superior to CpG alone at inducing regression of established tumours in several murine tumour models, primarily mediated by CD8+ T cells. An antitumour effect on distant untreated tumours (abscopal effect) was reliant on sustained activity of NKT cells and was associated with infiltration of KLRG1+ NKT cells in tumours and draining lymph nodes at both injected and untreated distant sites. Cytometric analysis pointed to increased exposure to type I interferon (IFN) affecting many immune cell types in the tumour and lymphoid organs. Accordingly, antitumour activity was lost in animals in which dendritic cells (DCs) were incapable of signaling through the type I IFN receptor. Studies in conditional ablation models showed that conventional type 1 DCs and plasmacytoid DCs were required for the response. In tumour models where the combined treatment was less effective, the addition of tumour-antigen derived peptide, preferably conjugated to α-GalCer, significantly enhanced the antitumour response. The combination of TLR ligation, NKT cell agonism, and peptide delivery could therefore be adapted to induce responses to both known and unknown antigens.
Collapse
Affiliation(s)
- Kef K Prasit
- Malaghan Institute of Medical Research, Wellington, New Zealand.,Maurice Wilkins Centre, Auckland, New Zealand
| | - Laura Ferrer-Font
- Maurice Wilkins Centre, Auckland, New Zealand.,Hugh Green Cytometry Centre, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Olivia K Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Regan J Anderson
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Benjamin J Compton
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Alfonso J Schmidt
- Hugh Green Cytometry Centre, Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Chun-Jen J Chen
- Maurice Wilkins Centre, Auckland, New Zealand.,School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | | | - David S Ritchie
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia.,University of Melbourne, Melbourne, Australia
| | - Dale I Godfrey
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Australia
| | - Stephen R Mattarollo
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - P Rod Dundar
- Maurice Wilkins Centre, Auckland, New Zealand.,School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand.,Maurice Wilkins Centre, Auckland, New Zealand
| |
Collapse
|
6
|
Zhao L, Yang X. Cross Talk Between Natural Killer T and Dendritic Cells and Its Impact on T Cell Responses in Infections. Front Immunol 2022; 13:837767. [PMID: 35185930 PMCID: PMC8850912 DOI: 10.3389/fimmu.2022.837767] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/19/2022] [Indexed: 11/16/2022] Open
Abstract
Both innate and adaptive immunity is vital for host defense against infections. Dendritic cells (DCs) are critical for initiating and modulating adaptive immunity, especially for T-cell responses. Natural killer T (NKT) cells are a small population of innate-like T cells distributed in multiple organs. Many studies have suggested that the cross-talk between these two immune cells is critical for immunobiology and host defense mechanisms. Not only can DCs influence the activation/function of NKT cells, but NKT cells can feedback on DCs also, thus modulating the phenotype and function of DCs and DC subsets. This functional feedback of NKT cells on DCs, especially the preferential promoting effect on CD8α+ and CD103+ DC subsets in lymphoid and non-lymphoid tissues, significantly impacts the systemic and local adaptive CD4 and CD8 T cell responses in infections. This review focuses on the two-way interaction between NKT cells and DCs, emphasizing the importance of NKT cell feedback on DCs in bridging innate and adaptive immune responses for host defense purposes.
Collapse
Affiliation(s)
- Lei Zhao
- Departments of Immunology and Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Xi Yang
- Departments of Immunology and Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
7
|
iNKT cell agonists as vaccine adjuvants to combat infectious diseases. Carbohydr Res 2022; 513:108527. [DOI: 10.1016/j.carres.2022.108527] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 01/07/2023]
|
8
|
Burn OK, Pankhurst TE, Painter GF, Connor LM, Hermans IF. Harnessing NKT cells for vaccination. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab013. [PMID: 36845569 PMCID: PMC9914585 DOI: 10.1093/oxfimm/iqab013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 11/14/2022] Open
Abstract
Natural killer T (NKT) cells are innate-like T cells capable of enhancing both innate and adaptive immune responses. When NKT cells are stimulated in close temporal association with co-administered antigens, strong antigen-specific immune responses can be induced, prompting the study of NKT cell agonists as novel immune adjuvants. This activity has been attributed to the capacity of activated NKT cells to act as universal helper cells, with the ability to provide molecular signals to dendritic cells and B cells that facilitate T cell and antibody responses, respectively. These signals can override the requirement for conventional CD4+ T cell help, so that vaccines can be designed without need to consider CD4+ T cell repertoire and major histocompatibility complex Class II diversity. Animal studies have highlighted some drawbacks of the approach, namely, concerns around induction of NKT cell hyporesponsiveness, which may limit vaccine boosting, and potential for toxicity. Here we highlight studies that suggest these obstacles can be overcome by targeted delivery in vivo. We also feature new studies that suggest activating NKT cells can help encourage differentiation of T cells into tissue-resident memory cells that play an important role in prophylaxis against infection, and may be required in cancer therapy.
Collapse
Affiliation(s)
- Olivia K Burn
- Malaghan Institute of Medical Research, PO Box 7060, Wellington 6042, New Zealand
| | - Theresa E Pankhurst
- The School of Biological Sciences, Victoria University of Wellington, PO Box 600, Wellington 6140, New Zealand
| | - Gavin F Painter
- The Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Petone 5046, New Zealand,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Lisa M Connor
- Malaghan Institute of Medical Research, PO Box 7060, Wellington 6042, New Zealand,The School of Biological Sciences, Victoria University of Wellington, PO Box 600, Wellington 6140, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, PO Box 7060, Wellington 6042, New Zealand,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland, New Zealand,Correspondence address. Malaghan Institute of Medical Research, Wellington, New Zealand. Tel: +64 4 4996914; E-mail: (I.F.H.)
| |
Collapse
|
9
|
Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology 2021; 1:894-907. [PMID: 23162757 PMCID: PMC3489745 DOI: 10.4161/onci.20931] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptors (TLRs) have first been characterized for their capacity to detect conserved microbial components like lipopolysaccharide (LPS) and double-stranded RNA, resulting in the elicitation of potent (innate) immune responses against invading pathogens. More recently, TLRs have also been shown to promote the activation of the cognate immune system against cancer cells. Today, only three TLR agonists are approved by FDA for use in humans: the bacillus Calmette-Guérin (BCG), monophosphoryl lipid A (MPL) and imiquimod. BCG (an attenuated strain of Mycobacterium bovis) is mainly used as a vaccine against tuberculosis, but also for the immunotherapy of in situ bladder carcinoma. MPL (derived from the LPS of Salmonella minnesota) is included in the formulation of Cervarix®, a vaccine against human papillomavirus-16 and -18. Imiquimod (a synthetic imidazoquinoline) is routinely employed for actinic keratosis, superficial basal cell carcinoma, and external genital warts (condylomata acuminata). In this Trial Watch, we will summarize the results of recently completed clinical trials and discuss the progress of ongoing studies that have evaluated/are evaluating FDA-approved TLR agonists as off-label medications for cancer therapy.
Collapse
Affiliation(s)
- Erika Vacchelli
- INSERM, U848; Villejuif, France ; Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Intranasal Therapeutic Peptide Vaccine Promotes Efficient Induction and Trafficking of Cytotoxic T Cell Response for the Clearance of HPV Vaginal Tumors. Vaccines (Basel) 2020; 8:vaccines8020259. [PMID: 32485935 PMCID: PMC7349944 DOI: 10.3390/vaccines8020259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/18/2020] [Accepted: 05/27/2020] [Indexed: 11/30/2022] Open
Abstract
Human papillomavirus (HPV)-induced cancers continue to affect millions of women around the world, and the five year survival rate under the current standard of care for these cancers is less than 60% in some demographics. Therefore there is still an unmet need to develop an effective therapy that can be easily administered to treat established HPV cervical cancer lesions. We sought to investigate the potential of an intranasal HPV peptide therapeutic vaccine incorporating the combination of α-Galactosylceramide (α-GalCer) and CpG-ODN adjuvants (TVAC) against established HPV genital tumors in a syngeneic C57BL/6J mouse model. We obtained evidence to show that TVAC, delivered by the mucosal intranasal route, induced high frequencies of antigen-specific CD8 T cells concurrent with significant reduction in the immunosuppressive regulatory T cells and myeloid derived suppressor cells in the tumor microenvironment (TME), correlating with sustained elimination of established HPV genital tumors in over 85% of mice. Inclusion of both the adjuvants in the vaccine was necessary for significant increase of antigen-specific CD8 T cells to the tumor and antitumor efficacy because vaccination incorporating either adjuvant alone was inefficient. These results strongly support the utility of the TVAC administered by needle-free intranasal route as a safe and effective strategy for the treatment of established genital HPV tumors.
Collapse
|
11
|
Guevara M, Jilesen Z, Stojdl D, Persano S. Codelivery of mRNA with α-Galactosylceramide Using a New Lipopolyplex Formulation Induces a Strong Antitumor Response upon Intravenous Administration. ACS OMEGA 2019; 4:13015-13026. [PMID: 31460428 PMCID: PMC6705043 DOI: 10.1021/acsomega.9b00489] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/23/2019] [Indexed: 05/22/2023]
Abstract
Recently, the use of mRNA-based vaccines for cancer immunotherapy has gained growing attention. Several studies have shown that mRNA delivered in a vectorized format can generate a robust and efficient immune response. In this work, a new lipopolyplex vector (multi-LP), incorporating the immune adjuvant α-galactosylceramide (α-GalCer) and a multivalent cationic lipid, was proposed for the in vivo delivery of mRNA into antigen-presenting cells. We demonstrate that dendritic cells (DCs) can be targeted in vivo by intravenous administration of a α-GalCer-/mRNA-loaded multi-LP vector, without the need for its functionalization with cell-specific antibodies or ligands. The multi-LP nanoparticles loaded with a reporter mRNA efficiently led to high expression of the enhanced green fluorescence protein in DCs both in vitro and in vivo, exhibiting an intrinsic selectivity for DCs. Finally, the TRP2-mRNA/α-GalCer-based multi-LP vaccine induced a significant therapeutic effect against a highly malignant B16-F10 melanoma tumor. This study provides the first evidence that a combination of antigen-mRNA and α-GalCer can be used as an effective antitumor vaccine, inducing strong innate and adaptive immune responses.
Collapse
Affiliation(s)
- Maria
L. Guevara
- Children’s
Hospital of Eastern Ontario (CHEO) Research Institute, Department
of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1N 6N5, Canada
| | - Zachary Jilesen
- Children’s
Hospital of Eastern Ontario (CHEO) Research Institute, Department
of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1N 6N5, Canada
| | - David Stojdl
- Children’s
Hospital of Eastern Ontario (CHEO) Research Institute, Department
of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1N 6N5, Canada
- E-mail: (D.S.)
| | - Stefano Persano
- Children’s
Hospital of Eastern Ontario (CHEO) Research Institute, Department
of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1N 6N5, Canada
- Istituto
Italiano di Tecnologia (IIT), Via Morego 30, Genova 16163, Italy
- E-mail: (S.P.)
| |
Collapse
|
12
|
Chen XZ, Zhang RY, Wang XF, Yin XG, Wang J, Wang YC, Liu X, Du JJ, Liu Z, Guo J. Peptide-free Synthetic Nicotine Vaccine Candidates with α-Galactosylceramide as Adjuvant. Mol Pharm 2019; 16:1467-1476. [DOI: 10.1021/acs.molpharmaceut.8b01095] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Xiang-Zhao Chen
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Ru-Yan Zhang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Xi-Feng Wang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Xu-Guang Yin
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Jian Wang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Ya-Cong Wang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Xiu Liu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Jing-Jing Du
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Zheng Liu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Jun Guo
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| |
Collapse
|
13
|
|
14
|
Weyant KB, Mills DC, DeLisa MP. Engineering a new generation of carbohydrate-based vaccines. Curr Opin Chem Eng 2018; 19:77-85. [PMID: 30568873 DOI: 10.1016/j.coche.2017.12.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Recent advances in chemical synthesis, conjugation chemistry, engineered biosynthesis, and formulation design have spawned a new generation of vaccines that incorporate carbohydrate antigens. By providing better immunity against a variety of pathogens or malignant cells and lowering the cost of production, these developments overcome many of the limitations associated with conventional vaccines involving polysaccharides. Moreover, the resulting vaccine candidates are shedding light on how the immune system responds to carbohydrates and providing mechanistic insight that can help guide future vaccine design. Here, we review recent engineering efforts to develop and manufacture carbohydrate-based vaccines that are efficacious, durable, and cost-effective.
Collapse
Affiliation(s)
- Kevin B Weyant
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853 USA
| | - Dominic C Mills
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853 USA
| | - Matthew P DeLisa
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853 USA.,Comparative Biomedical Sciences, Cornell University, Ithaca, NY 14853 USA.,Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853 USA
| |
Collapse
|
15
|
He Q, Liu L, Yang Q, Wang A, Chen S, Li R, Huang Y, Zhang G, Ding X, Yu H, Hu S, Nie H. Invariant natural killer T cells promote immunogenic maturation of lung dendritic cells in mouse models of asthma. Am J Physiol Lung Cell Mol Physiol 2017; 313:L973-L990. [PMID: 28912381 DOI: 10.1152/ajplung.00340.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 01/17/2023] Open
Abstract
Our previous study showed that invariant natural killer T (iNKT) cells might act as an adjuvant to promote Th2 inflammatory responses in an OVA-induced mouse model of allergic asthma, but the mechanism remains unknown. To clarify the underlying mechanism through which iNKT cells promote Th2 inflammatory responses, we investigated the modulatory influence of iNKT cells on phenotypic and functional maturation of lung dendritic cells (LDCs) using iNKT cell-knockout mice, specific iNKT cell activation, coculture experiments, and adoptive transfer of iNKT cells in mouse models of asthma. Our data showed that iNKT cell deficiency could downregulate surface maturation markers and proinflammatory cytokine secretion of LDCs from a mouse model of asthma. However, elevated activation of iNKT cells by α-galactosylceramide and adoptive transfer of iNKT cells could upregulate surface maturation markers and proinflammatory cytokine secretion of LDCs from mouse models of asthma. Meanwhile, iNKT cells significantly influenced the function of LDCs, markedly enhancing Th2 responses in vivo and in vitro. In addition, iNKT cell can induce LDCs expression of CD206 and RELM-α, reflecting alternative activation of LDCs in a mouse model of asthma. α-Galactosylceramide treatment significantly enhanced expression of CD40L of lung iNKT cells from a mouse model of asthma, and the coculture experiment of LDCs with iNKT cells showed that the blockade of CD40L strongly suppressed surface maturation markers and proinflammatory cytokine production by LDCs. Our data suggest that iNKT cells can promote immunogenic maturation of LDCs to enhance Th2 responses in mouse models of asthma.
Collapse
Affiliation(s)
- Qing He
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Linlin Liu
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qiaoyu Yang
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ailing Wang
- Nursing Department, Wuhan University School of Health Sciences, Wuhan, China; and
| | - Shuo Chen
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruiyun Li
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Huang
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guqin Zhang
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuhong Ding
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongying Yu
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Suping Hu
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hanxiang Nie
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China;
| |
Collapse
|
16
|
Ghinnagow R, De Meester J, Cruz LJ, Aspord C, Corgnac S, Macho-Fernandez E, Soulard D, Fontaine J, Chaperot L, Charles J, Soncin F, Mami-Chouaib F, Plumas J, Faveeuw C, Trottein F. Co-delivery of the NKT agonist α-galactosylceramide and tumor antigens to cross-priming dendritic cells breaks tolerance to self-antigens and promotes antitumor responses. Oncoimmunology 2017; 6:e1339855. [PMID: 28932640 DOI: 10.1080/2162402x.2017.1339855] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 06/01/2017] [Accepted: 06/01/2017] [Indexed: 10/19/2022] Open
Abstract
Vaccines designed to abrogate the tolerance of tumor self-antigens and amplify cytotoxic CD8+ T cells (CTLs) have promise for the treatment of cancer. Type I natural killer (NKT) cells have attracted considerable interest in the cancer therapy field. In the current study, we have exploited the unique ability of NKT cells to serve as T-helper cells to license dendritic cells (DCs) for cross priming with the aim to generate efficient CTL antitumor responses. To this end, we designed a nanoparticle-based vaccine to target cross-priming DCs via the Clec9a endocytic pathway. Our results showed for the first time that simultaneous co-delivery of the NKT agonist α-galactosylceramide and tumor self-antigens (Trp2 and gp100) to CD8α+ DCs promotes strong antitumor responses in prophylactic and therapeutic settings (advanced solid tumor model in the mouse). We attributed the vaccine's therapeutic effects to NKT cells (but not to T-helper lymphocytes) and CD8+ T cells. Efficacy was correlated with an elevated ratio between tumor antigen-specific CD8+ T cells and regulatory CD4+ T lymphocytes within the tumor. The nanoparticle-based vaccine actively targeted human CLEC9A-expressing BDCA3+ DCs - the equivalent of murine cross-priming CD8α+ DCs - and induced a strong expansion of effector memory tumor self-antigen (Melan -A)-specific CD8+ T cells from peripheral blood mononuclear cells sourced from healthy donors and melanoma patients. Together, our result shed light on novel therapeutic approaches for controlling tumor development.
Collapse
Affiliation(s)
- Reem Ghinnagow
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| | - Julie De Meester
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| | - Luis Javier Cruz
- Translational Nanobiomaterials and Imaging, Department of radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Caroline Aspord
- Institute for Advanced Biosciences (IAB), Team Immunobiology and Immunotherapy in Chronic Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Etablissement Français du Sang-Rhone-Alpes, F-38700 Grenoble, France
| | - Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, Fac. de médecine - Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Elodie Macho-Fernandez
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| | - Daphnée Soulard
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| | - Josette Fontaine
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| | - Laurence Chaperot
- Institute for Advanced Biosciences (IAB), Team Immunobiology and Immunotherapy in Chronic Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Etablissement Français du Sang-Rhone-Alpes, F-38700 Grenoble, France
| | - Julie Charles
- Institute for Advanced Biosciences (IAB), Team Immunobiology and Immunotherapy in Chronic Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Etablissement Français du Sang-Rhone-Alpes, F-38700 Grenoble, France.,Dermatology department, Grenoble Alpes University Hospital, F-38700 Grenoble, France
| | - Fabrice Soncin
- Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8161, F-59000 Lille, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, Fac. de médecine - Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Joel Plumas
- Institute for Advanced Biosciences (IAB), Team Immunobiology and Immunotherapy in Chronic Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Etablissement Français du Sang-Rhone-Alpes, F-38700 Grenoble, France
| | - Christelle Faveeuw
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| | - François Trottein
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| |
Collapse
|
17
|
Ghinnagow R, Cruz LJ, Macho-Fernandez E, Faveeuw C, Trottein F. Enhancement of Adjuvant Functions of Natural Killer T Cells Using Nanovector Delivery Systems: Application in Anticancer Immune Therapy. Front Immunol 2017; 8:879. [PMID: 28798749 PMCID: PMC5529346 DOI: 10.3389/fimmu.2017.00879] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/10/2017] [Indexed: 11/13/2022] Open
Abstract
Type I natural killer T (NKT) cells have gained considerable interest in anticancer immune therapy over the last decade. This “innate-like” T lymphocyte subset has the unique ability to recognize foreign and self-derived glycolipid antigens in association with the CD1d molecule expressed by antigen-presenting cells. An important property of these cells is to bridge innate and acquired immune responses. The adjuvant function of NKT cells might be exploited in the clinics. In this review, we discuss the approaches currently being used to target NKT cells for cancer therapy. In particular, we highlight ongoing strategies utilizing NKT cell-based nanovaccines to optimize immune therapy.
Collapse
Affiliation(s)
- Reem Ghinnagow
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - Luis Javier Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Elodie Macho-Fernandez
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - Christelle Faveeuw
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - François Trottein
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| |
Collapse
|
18
|
Speir M, Hermans IF, Weinkove R. Engaging Natural Killer T Cells as 'Universal Helpers' for Vaccination. Drugs 2017; 77:1-15. [PMID: 28005229 DOI: 10.1007/s40265-016-0675-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Conventional vaccine adjuvants enhance peptide-specific T-cell and B-cell responses by modifying peptide stability or uptake or by binding to pattern-recognition receptors on antigen-presenting cells (APCs). This article discusses the application of a distinct mechanism of adjuvant activity: the activation of type I, or invariant, natural killer T (iNKT) cells to drive cellular and humoral immune responses. Using a semi-invariant T-cell receptor (TCR), iNKT cells recognize glycolipid antigens presented on cluster of differentiation (CD)-1d molecules. When their ligands are presented in concert with peptides, iNKT cells can provide T-cell help, 'licensing' APCs to augment peptide-specific T-cell and antibody responses. We discuss the potential benefits and limitations of exploiting iNKT cells as 'universal helpers' to enhance vaccine responses for the treatment and prevention of cancer and infectious diseases.
Collapse
Affiliation(s)
- Mary Speir
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand. .,School of Biological Sciences, Victoria University Wellington, PO Box 600, Wellington, 6140, New Zealand. .,Maurice Wilkins Centre, Private Bag 92019, Auckland, New Zealand.
| | - Robert Weinkove
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand. .,Wellington Blood and Cancer Centre, Wellington Hospital, Private Bag 7902, Wellington, 6242, New Zealand. .,Department of Pathology and Molecular Medicine, University of Otago Wellington, Wellington, 6021, New Zealand.
| |
Collapse
|
19
|
Polonskaya Z, Deng S, Sarkar A, Kain L, Comellas-Aragones M, McKay CS, Kaczanowska K, Holt M, McBride R, Palomo V, Self KM, Taylor S, Irimia A, Mehta SR, Dan JM, Brigger M, Crotty S, Schoenberger SP, Paulson JC, Wilson IA, Savage PB, Finn MG, Teyton L. T cells control the generation of nanomolar-affinity anti-glycan antibodies. J Clin Invest 2017; 127:1491-1504. [PMID: 28287405 DOI: 10.1172/jci91192] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/19/2017] [Indexed: 12/27/2022] Open
Abstract
Vaccines targeting glycan structures at the surface of pathogenic microbes must overcome the inherent T cell-independent nature of immune responses against glycans. Carbohydrate conjugate vaccines achieve this by coupling bacterial polysaccharides to a carrier protein that recruits heterologous CD4 T cells to help B cell maturation. Yet they most often produce low- to medium-affinity immune responses of limited duration in immunologically fit individuals and disappointing results in the elderly and immunocompromised patients. Here, we hypothesized that these limitations result from suboptimal T cell help. To produce the next generation of more efficacious conjugate vaccines, we have explored a synthetic design aimed at focusing both B cell and T cell recognition to a single short glycan displayed at the surface of a virus-like particle. We tested and established the proof of concept of this approach for 2 serotypes of Streptococcus pneumoniae. In both cases, these vaccines elicited serotype-specific, protective, and long-lasting IgG antibodies of nanomolar affinity against the target glycans in mice. We further identified a requirement for CD4 T cells in the anti-glycan antibody response. Our findings establish the design principles for improved glycan conjugate vaccines. We surmise that the same approach can be used for any microbial glycan of interest.
Collapse
MESH Headings
- Adult
- Amino Acid Sequence
- Animals
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/chemistry
- Antibody Affinity
- B-Lymphocytes/immunology
- Bacterial Proteins/immunology
- CD4-Positive T-Lymphocytes/immunology
- Child
- Crystallography, X-Ray
- Female
- Glycopeptides/immunology
- Humans
- Hybridomas
- Immunoglobulin G/blood
- Male
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Models, Molecular
- Pneumococcal Infections/immunology
- Pneumococcal Infections/prevention & control
- Pneumococcal Vaccines/chemistry
- Pneumococcal Vaccines/immunology
- Polysaccharides, Bacterial/chemistry
- Polysaccharides, Bacterial/immunology
- Protein Binding
- Streptococcus pneumoniae/immunology
- Vaccination
- Vaccine Potency
Collapse
|
20
|
Lee HW, Jie HB, Bollyky PL, Sarracino D, Kim TS, Wilson BS. Role of dendritic cell maturation factors produced by human invariant NKT cells in immune tolerance. J Leukoc Biol 2016; 101:989-1003. [PMID: 27837018 DOI: 10.1189/jlb.1a0416-164rrr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 10/04/2016] [Accepted: 10/27/2016] [Indexed: 01/12/2023] Open
Abstract
In this study, we used the culture supernatant of iNKT cells to identify human myeloid DC maturation factors produced by human CD4+ iNKT cells. S100A8 had a strong maturation effect. Notably, the recombinant S100A8 protein displayed properties of DC maturation functioning, and the induction of DC differentiation by both the purified and the recombinant protein were blocked by anti-S100A8 and anti-TLR-4 mAbs. DC differentiation induced by anti-major histocompatibility complex class II/CD1d Ab, S100A8, or both was qualitatively indistinguishable from that induced by the coculture of DCs and iNKT cells or via culture supplementation with supernatants from activated CD4+ iNKT cells. S100A8 also induced CD4+/CD25+/Foxp3+ Treg cells from naïve T cells. S100A8 may contribute to DC differentiation by elevating transcription factors or activating transcription factor-2, heat shock factor-1, or both, in mature DCs. S100A8 is a novel candidate iNKT cell-dependent DC maturation factor.
Collapse
Affiliation(s)
- Hyeong-Woo Lee
- Departments of Tropical Medicine and Parasitology, Inha University School of Medicine, Incheon, Republic of Korea.,Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Hyun Bae Jie
- OncoMed Pharmaceuticals, Inc., Redwood City, California
| | - Paul L Bollyky
- Division of Infectious Diseases, Stanford University Medical Center, Stanford, California; and
| | - David Sarracino
- Thermo Fisher Scientific Biomarkers Research Initiatives in Mass Spectrometry (BRIMS) Center, Cambridge, Massachusetts
| | - Tong-Soo Kim
- Departments of Tropical Medicine and Parasitology, Inha University School of Medicine, Incheon, Republic of Korea;
| | - Brian S Wilson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida;
| |
Collapse
|
21
|
Immunotherapeutic strategies targeting natural killer T cell responses in cancer. Immunogenetics 2016; 68:623-38. [PMID: 27393665 DOI: 10.1007/s00251-016-0928-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/22/2016] [Indexed: 12/21/2022]
Abstract
Natural killer T (NKT) cells are a unique subset of lymphocytes that bridge the innate and adaptive immune system. NKT cells possess a classic αβ T cell receptor (TCR) that is able to recognize self and foreign glycolipid antigens presented by the nonclassical class I major histocompatibility complex (MHC) molecule, CD1d. Type I NKT cells (referred to as invariant NKT cells) express a semi-invariant Vα14Jα18 TCR in mice and Vα24Jα18 TCR in humans. Type II NKT cells are CD1d-restricted T cells that express a more diverse set of TCR α chains. The two types of NKT cells often exert opposing effects especially in tumor immunity, where type II cells generally suppress tumor immunity while type I NKT cells can enhance anti-tumor immune responses. In this review, we focus on the role of NKT cells in cancer. We discuss their effector and suppressive functions, as well as describe preclinical and clinical studies utilizing therapeutic strategies focused on harnessing their potent anti-tumor effector functions, and conclude with a discussion on potential next steps for the utilization of NKT cell-targeted therapies for the treatment of cancer.
Collapse
|
22
|
Gableh F, Saeidi M, Hemati S, Hamdi K, Soleimanjahi H, Gorji A, Ghaemi A. Combination of the toll like receptor agonist and α-Galactosylceramide as an efficient adjuvant for cancer vaccine. J Biomed Sci 2016; 23:16. [PMID: 26811064 PMCID: PMC4727273 DOI: 10.1186/s12929-016-0238-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 01/20/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND DNA vaccines have emerged as an attractive approach for the generation of cytotoxic T lymphocytes (CTL). In our previous study, we found That Toll like receptor (TLR) ligands are promising candidates for the development of novel adjuvants for DNA vaccine. To improve the efficacy of DNA vaccine directed against human papillomavirus (HPV) tumors, we evaluated whether co-administration of a TLR4 ligand, monophosphoryl lipid A (MPL), and Natural Killer T Cell Ligand α-Galactosylceramide(α-GalCer) adjuvants with DNA vaccine would influence the anti-tumor efficacy of DNA vaccinations. METHODS We investigated the effectiveness of α-GalCer and MPL combination as an adjuvant with an HPV-16 E7 DNA vaccine to enhance antitumor immune responses. RESULTS By using adjuvant combination for a DNA vaccine, we found that the levels of lymphocyte proliferation, CTL activity, IFN- γ, IL-4 and IL-12 responses, and tumor protection against TC-1 cells were significantly increased compared to the DNA vaccine with individual adjuvants. In addition, inhibition of IL-18 signaling during vaccination decreased IFN-γ responses and tumor protection, and that this inhibition suggested stimulatory role of IL-18 in adjuvant effects of α-GalCer and MPL combination. CONCLUSION The strong adjuvanticity associated with α-GalCer/MPL combination may to be an important tool in the development of novel and strong cancer immunotherapy.
Collapse
Affiliation(s)
- Fateme Gableh
- Infectious Diseases Research Center, Department of Microbiology, Golestan University of Medical Sciences, POBox: 49175-1141, Gorgan, Iran
| | - Mohsen Saeidi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shaghayegh Hemati
- Guilan Science and Research Branch, Islamic Azad University, Rasht, Iran
| | - Kasra Hamdi
- Department of microbiology, Islamic Azad University, Shiraz branch, Shiraz, Iran
| | - Hoorieh Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Tehran, Iran.,Epilepsy Research Center, Institut für Physiologie I, Westfälische Wilhelms-Universität Münster, Robert-Koch-Strasse, Münster, Germany.,Klinik und Poliklinik für Neurochirurgie, Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Amir Ghaemi
- Infectious Diseases Research Center, Department of Microbiology, Golestan University of Medical Sciences, POBox: 49175-1141, Gorgan, Iran. .,Shefa Neuroscience Research Center, Tehran, Iran. .,Department of Virology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
23
|
Immunoregulation of NKT Cells in Systemic Lupus Erythematosus. J Immunol Res 2015; 2015:206731. [PMID: 26819956 PMCID: PMC4706917 DOI: 10.1155/2015/206731] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/02/2015] [Indexed: 01/17/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease with different variety of clinical manifestations. Natural killer T (NKT) cells are innate lymphocytes that play a regulatory role during broad range of immune responses. A number of studies demonstrated that the quantity and quality of invariant NKT (iNKT) cells showed marked defects in SLE patients in comparison to healthy controls. This finding suggests that iNKT cells may play a regulatory role in the occurrence and development of this disease. In this review, we mainly summarized the most recent findings about the behavior of NKT cells in SLE patients and mouse models, as well as how NKT cells affect the proportion of T helper cells and the production of autoreactive antibodies in the progress of SLE. This will help people better understand the role of NKT cells in the development of SLE and improve the therapy strategy.
Collapse
|
24
|
Co-operation of α-galactosylceramide-loaded tumour cells and TLR9 agonists induce potent anti-tumour responses in a murine colon cancer model. Biochem J 2015; 473:7-19. [PMID: 26450924 DOI: 10.1042/bj20150129] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 10/08/2015] [Indexed: 01/08/2023]
Abstract
Dendritic cells (DCs) and invariant natural killer T (iNKT) cells play important roles in linking innate immunity and adaptive immunity. Mature DCs activated by Toll-like receptor (TLR) agonists directly activate iNKT cells and the iNKT ligand α-galactosylceramide (α-Galcer) can induce DC maturation, resulting in enhanced protective immune responses. In the present study, we aimed to boost anti-tumour immunity in a murine colon cancer model by synergizing DCs and iNKT cells using α-Galcer-loaded tumour cells (tumour-Gal) and the TLR9 agonist cytosine-phosphorothioate-guanine (CpG1826). The vaccine strategy was sufficient to inhibit growth of established tumours and prolonged survival of tumour-bearing mice. Importantly, the immunization induced an adaptive memory immune response as the survivors from primary tumour inoculations were resistant to a tumour re-challenge. Furthermore, injection of tumour-Gal with CpG1826 resulted in iNKT cell activation and DC maturation as defined by interferon (IFN)-γ secretion by iNKT, natural killer (NK) cells and interleukin (IL)-12 by DCs. Immunohistochemistry analysis revealed that cluster of differentiation (CD)4(+) T-cells and CD8(+) T-cells played important roles in anti-tumour immunity. Additionally, the vaccine redirected Th2 (T-helper cell type 2) responses toward Th1 (T-helper cell type 1) responses with increases in IL-2, IFN-γ expression and decreases in IL-4 and IL-5 expression after immunization with tumour-Gal with CpG1826. Taken together, our results demonstrated a novel vaccination by synergizing tumour-Gal and CpG1826 against murine colon cancer, which can be further developed as tumour-specific immunotherapy against human cancer.
Collapse
|
25
|
Osmond TL, Farrand KJ, Painter GF, Ruedl C, Petersen TR, Hermans IF. Activated NKT Cells Can Condition Different Splenic Dendritic Cell Subsets To Respond More Effectively to TLR Engagement and Enhance Cross-Priming. THE JOURNAL OF IMMUNOLOGY 2015; 195:821-31. [PMID: 26078270 DOI: 10.4049/jimmunol.1401751] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 05/20/2015] [Indexed: 11/19/2022]
Abstract
The function of dendritic cells (DCs) can be modulated through multiple signals, including recognition of pathogen-associated molecular patterns, as well as signals provided by rapidly activated leukocytes in the local environment, such as innate-like T cells. In this article, we addressed the possibility that the roles of different murine DC subsets in cross-priming CD8(+) T cells can change with the nature and timing of activatory stimuli. We show that CD8α(+) DCs play a critical role in cross-priming CD8(+) T cell responses to circulating proteins that enter the spleen in close temporal association with ligands for TLRs and/or compounds that activate NKT cells. However, if NKT cells are activated first, then CD8α(-) DCs become conditioned to respond more vigorously to TLR ligation, and if triggered directly, these cells can also contribute to priming of CD8(+) T cell responses. In fact, the initial activation of NKT cells can condition multiple DC subsets to respond more effectively to TLR ligation, with plasmacytoid DCs making more IFN-α and both CD8α(+) and CD8α(-) DCs manufacturing more IL-12. These results suggest that different DC subsets can contribute to T cell priming if provided appropriately phased activatory stimuli, an observation that could be factored into the design of more effective vaccines.
Collapse
Affiliation(s)
- Taryn L Osmond
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand; School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Kathryn J Farrand
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Gavin F Painter
- The Ferrier Research Institute, Victoria University of Wellington, Lower Hutt 5010, New Zealand; and
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Troels R Petersen
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand; School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand;
| |
Collapse
|
26
|
Corgnac S, Perret R, Zhang L, Mach JP, Romero P, Donda A. iNKT/CD1d-antitumor immunotherapy significantly increases the efficacy of therapeutic CpG/peptide-based cancer vaccine. J Immunother Cancer 2014; 2:39. [PMID: 25426294 PMCID: PMC4243737 DOI: 10.1186/s40425-014-0039-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 10/08/2014] [Indexed: 11/26/2022] Open
Abstract
Background Therapeutic cancer vaccines aim to boost the natural immunity against transformed cancer cells, and a series of adjuvants and co-stimulatory molecules have been proposed to enhance the immune response against weak self-antigens expressed on cancer cells. For instance, a peptide/CpG-based cancer vaccine has been evaluated in several clinical trials and was shown in pre-clinical studies to favor the expansion of effector T versus Tregs cells, resulting in a potent antitumor activity, as compared to other TLR ligands. Alternatively, the adjuvant activity of CD1d-restricted invariant NKT cells (iNKT) on the innate and adaptive immunity is well demonstrated, and several CD1d glycolipid ligands are under pre-clinical and clinical evaluation. Importantly, additive or even synergistic effects have been shown upon combined CD1d/NKT agonists and TLR ligands. The aim of the present study is to combine the activation and tumor targeting of activated iNKT, NK and T cells. Methods Activation and tumor targeting of iNKT cells via recombinant α-galactosylceramide (αGC)-loaded CD1d-anti-HER2 fusion protein (CD1d-antitumor) is combined or not with OVA peptide/CpG vaccine. Circulating and intratumoral NK and H-2Kb/OVA-specific CD8 responses are monitored, as well as the state of activation of dendritic cells (DC) with regard to activation markers and IL-12 secretion. The resulting antitumor therapy is tested against established tumor grafts of B16 melanoma cells expressing human HER2 and ovalbumin. Results The combined CD1d/iNKT antitumor therapy and CpG/peptide-based immunization leads to optimized expansion of NK and OVA-specific CD8 T cells (CTLs), likely resulting from the maturation of highly pro-inflammatory DCs as seen by a synergistic increase in serum IL-12. The enhanced innate and adaptive immune responses result in higher tumor inhibition that correlates with increased numbers of OVA-specific CTLs at the tumor site. Antibody-mediated depletion experiments further demonstrate that in this context, CTLs rather than NK cells are essential for the enhanced tumor inhibition. Conclusions Altogether, our study in mice demonstrates that αGC/CD1d-antitumor fusion protein greatly increases the efficacy of a therapeutic CpG-based cancer vaccine, first as an adjuvant during T cell priming and second, as a therapeutic agent to redirect immune responses to the tumor site. Electronic supplementary material The online version of this article (doi:10.1186/s40425-014-0039-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stéphanie Corgnac
- Translational Tumor Immunology Group, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Rachel Perret
- Translational Tumor Immunology Group, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Lianjun Zhang
- Translational Tumor Immunology Group, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Jean-Pierre Mach
- Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Pedro Romero
- Translational Tumor Immunology Group, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Alena Donda
- Translational Tumor Immunology Group, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
27
|
An autologous leukemia cell vaccine prevents murine acute leukemia relapse after cytarabine treatment. Blood 2014; 124:2953-63. [PMID: 25237205 DOI: 10.1182/blood-2014-04-568956] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Acute leukemias with adverse prognostic features carry a high relapse rate without allogeneic stem cell transplantation (allo-SCT). Allo-SCT has a high morbidity and is precluded for many patients because of advanced age or comorbidities. Postremission therapies with reduced toxicities are urgently needed. The murine acute leukemia model C1498 was used to study the efficacy of an intravenously administered vaccine consisting of irradiated leukemia cells loaded with the natural killer T (NKT)-cell agonist α-galactosylceramide (α-GalCer). Prophylactically, the vaccine was highly effective at preventing leukemia development through the downstream activities of activated NKT cells, which were dependent on splenic langerin(+)CD8α(+) dendritic cells and which led to stimulation of antileukemia CD4(+) and CD8(+) T cells. However, hosts with established leukemia received no protective benefit from the vaccine, despite inducing NKT-cell activation. Established leukemia was associated with increases in regulatory T cells and myeloid-derived suppressor cells, and the leukemic cells themselves were highly suppressive in vitro. Although this suppressive environment impaired both effector arms of the immune response, CD4(+) T-cell responses were more severely affected. When cytarabine chemotherapy was administered prior to vaccination, all animals in remission posttherapy were protected against rechallenge with viable leukemia cells.
Collapse
|
28
|
Shekhar S, Joyee AG, Yang X. Invariant natural killer T cells: boon or bane in immunity to intracellular bacterial infections? J Innate Immun 2014; 6:575-84. [PMID: 24903638 DOI: 10.1159/000361048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 03/04/2014] [Indexed: 11/19/2022] Open
Abstract
Invariant natural killer T (iNKT) cells represent a specialized subset of innate lymphocytes that recognize lipid and glycolipid antigens presented to them by nonclassical MHC-I CD1d molecules and are able to rapidly secrete copious amounts of a variety of cytokines. iNKT cells possess the ability to modulate innate as well as adaptive immune responses against various pathogens. Intracellular bacteria are one of the most clinically significant human pathogens that effectively evade the immune system and cause a myriad of diseases of public health concern globally. Emerging evidence suggests that iNKT cells can confer immunity to intracellular bacteria but also inflict pathology in certain cases. We summarize the current knowledge on the contribution of iNKT cells in the host defense against intracellular bacterial infections, with a focus on the underlying mechanisms by which these cells induce protective or pathogenic reactions including the pathways of direct action (acting on infected cells) and indirect action (modulating dendritic, NK and T cells). The rational exploitation of iNKT cells for prophylactic and therapeutic purposes awaits a profound understanding of their functional biology.
Collapse
Affiliation(s)
- Sudhanshu Shekhar
- Laboratory for Infection and Immunity, Department of Medical Microbiology, University of Manitoba, Winnipeg, Man., Canada
| | | | | |
Collapse
|
29
|
Villanueva AI, Haeryfar SMM, Mallard BA, Kulkarni RR, Sharif S. Functions of invariant NK T cells are modulated by TLR ligands and IFN-α. Innate Immun 2014; 21:275-88. [PMID: 24934453 DOI: 10.1177/1753425914527327] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Invariant NK T (iNKT) cells perform numerous immunoregulatory functions. In mice, they express a unique and invariant Vα14-Jα18 rearrangement of α chain in their TCR recognizing glycolipid Ags presented by CD1d. This recognition results in the rapid release of both Th1- and Th2-type cytokines, making them early mediators of the immune response. Owing to their rapid activation and genetic rigidity of their TCR, iNKT cells share characteristics with innate lymphocytes. Therefore, we investigated whether iNKT cells could be induced to express TLRs, a class of pathogen recognition receptor. Mouse iNKT cells were stimulated with anti-CD3 monoclonal Ab and IFN-α, resulting in an increase in the transcription of TLRs 3, 5, 7 and 9, and increased surface expression of TLR3. These cells were subsequently stimulated with TLR ligands, resulting in an increase in the production of IFN-γ, IL-4 and TNF-α. Supernatants from these cells also increased macrophage production of IL-6 and prostaglandin E2, and increased their phagocytic activity and CD80 expression. These supernatants also reduced vesicular stomatitis virus-GFP replication in fibroblasts. This study demonstrates the role of IFN-α in iNKT cell activation, as well as the direct modulatory effects of TLR ligands on iNKT cell function, including antiviral activity.
Collapse
Affiliation(s)
- A Ian Villanueva
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Bonnie A Mallard
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Raveendra R Kulkarni
- Department of Pediatrics and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
30
|
Abstract
Over the past 15 years, investigators have shown that T lymphocytes can recognize not only peptides in the context of MHC class I and class II molecules but also foreign and self-lipids in association with the nonclassical MHC class I-like molecules, CD1 proteins. In this review, we describe the most recent events in the field, with particular emphasis on (a) structural and functional aspects of lipid presentation by CD1 molecules, (b) the development of CD1d-restricted invariant natural killer T (iNKT) cells and transcription factors required for their differentiation, (c) the ability of iNKT cells to modulate innate and adaptive immune responses through their cross talk with lymphoid and myeloid cells, and (d) MR1-restricted and group I (CD1a, CD1b, and CD1c)-restricted T cells.
Collapse
Affiliation(s)
- Mariolina Salio
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DU, United Kingdom;
| | | | | | | |
Collapse
|
31
|
Archer LD, Langford-Smith KJ, Bigger BW, Fildes JE. Mucopolysaccharide diseases: a complex interplay between neuroinflammation, microglial activation and adaptive immunity. J Inherit Metab Dis 2014; 37:1-12. [PMID: 23653226 DOI: 10.1007/s10545-013-9613-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 04/16/2013] [Indexed: 12/23/2022]
Abstract
Mucopolysaccharide (MPS) diseases are lysosomal storage disorders (LSDs) caused by deficiencies in enzymes required for glycosaminoglycan (GAG) catabolism. Mucopolysaccharidosis I (MPS I), MPS IIIA, MPS IIIB and MPS VII are deficient in the enzymes α-L-Iduronidase, Heparan-N-Sulphatase, N-Acetylglucosaminidase and Beta-Glucuronidase, respectively. Enzyme deficiency leads to the progressive multi-systemic build-up of heparan sulphate (HS) and dermatan sulphate (DS) within cellular lysosomes, followed by cell, tissue and organ damage and in particular neurodegeneration. Clinical manifestations of MPS are well established; however as lysosomes represent vital components of immune cells, it follows that lysosomal accumulation of GAGs could affect diverse immune functions and therefore influence disease pathogenesis. Theoretically, MPS neurodegeneration and GAGs could be substantiating a threat of danger and damage to alert the immune system for cellular clearance, which due to the progressive nature of MPS storage would propagate disease pathogenesis. Innate immunity appears to have a key role in MPS; however the extent of adaptive immune involvement remains to be elucidated. The current literature suggests a complex interplay between neuroinflammation, microglial activation and adaptive immunity in MPS disease.
Collapse
Affiliation(s)
- Louise D Archer
- The Transplant Centre, UHSM, University of Manchester, Manchester, England, UK
| | | | | | | |
Collapse
|
32
|
Dowds CM, Kornell SC, Blumberg RS, Zeissig S. Lipid antigens in immunity. Biol Chem 2014; 395:61-81. [PMID: 23999493 PMCID: PMC4128234 DOI: 10.1515/hsz-2013-0220] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 08/27/2013] [Indexed: 02/07/2023]
Abstract
Lipids are not only a central part of human metabolism but also play diverse and critical roles in the immune system. As such, they can act as ligands of lipid-activated nuclear receptors, control inflammatory signaling through bioactive lipids such as prostaglandins, leukotrienes, lipoxins, resolvins, and protectins, and modulate immunity as intracellular phospholipid- or sphingolipid-derived signaling mediators. In addition, lipids can serve as antigens and regulate immunity through the activation of lipid-reactive T cells, which is the topic of this review. We will provide an overview of the mechanisms of lipid antigen presentation, the biology of lipid-reactive T cells, and their contribution to immunity.
Collapse
Affiliation(s)
- C. Marie Dowds
- Department of Internal Medicine I, University Medical Center
Schleswig-Holstein, Schittenhelmstraße 12, D-24105 Kiel,
Germany
| | - Sabin-Christin Kornell
- Department of Internal Medicine I, University Medical Center
Schleswig-Holstein, Schittenhelmstraße 12, D-24105 Kiel,
Germany
| | - Richard S. Blumberg
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham
and Women’s Hospital, Harvard Medical School, 75 Francis Street,
Boston, MA 02115, USA
| | - Sebastian Zeissig
- Department of Internal Medicine I, University Medical Center
Schleswig-Holstein, Schittenhelmstraße 12, D-24105 Kiel,
Germany
| |
Collapse
|
33
|
Duman M, Chtcheglova LA, Zhu R, Bozna BL, Polzella P, Cerundolo V, Hinterdorfer P. Nanomapping of CD1d-glycolipid complexes on THP1 cells by using simultaneous topography and recognition imaging. J Mol Recognit 2013; 26:408-14. [DOI: 10.1002/jmr.2282] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/18/2013] [Accepted: 04/23/2013] [Indexed: 01/24/2023]
Affiliation(s)
| | | | - Rong Zhu
- Institute for Biophysics; University of Linz; Altenbergerstrasse 69; A-4040; Linz; Austria
| | - Bianca L. Bozna
- Institute for Biophysics; University of Linz; Altenbergerstrasse 69; A-4040; Linz; Austria
| | - Paolo Polzella
- Cancer Research UK Tumor Immunology Group, The Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine; University of Oxford; Oxford; OX3 9DS; UK
| | - Vicenzo Cerundolo
- Cancer Research UK Tumor Immunology Group, The Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine; University of Oxford; Oxford; OX3 9DS; UK
| | | |
Collapse
|
34
|
Karmakar S, Bhaumik SK, Paul J, De T. TLR4 and NKT cell synergy in immunotherapy against visceral leishmaniasis. PLoS Pathog 2012; 8:e1002646. [PMID: 22511870 PMCID: PMC3325212 DOI: 10.1371/journal.ppat.1002646] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 03/01/2012] [Indexed: 02/06/2023] Open
Abstract
NKT cells play an important role in autoimmune diseases, tumor surveillance, and infectious diseases, providing in most cases protection against infection. NKT cells are reactive to CD1d presented glycolipid antigens. They can modulate immune responses by promoting the secretion of type 1, type 2, or immune regulatory cytokines. Pathogen-derived signals to dendritic cells mediated via Toll like Receptors (TLR) can be modulated by activated invariant Natural Killer T (iNKT) cells. The terminal β-(1–4)-galactose residues of glycans can modulate host responsiveness in a T helper type-1 direction via IFN-γ and TLRs. We have attempted to develop a defined immunotherapeutic, based on the cooperative action of a TLR ligand and iNKT cell using a mouse model of visceral leishmaniasis. We evaluated the anti-Leishmania immune responses and the protective efficacy of the β-(1–4)-galactose terminal NKT cell ligand glycosphingophospholipid (GSPL) antigen of L. donovani parasites. Our results suggest that TLR4 can function as an upstream sensor for GSPL and provoke intracellular inflammatory signaling necessary for parasite killing. Treatment with GSPL was able to induce a strong effective T cell response that contributed to effective control of acute parasite burden and led to undetectable parasite persistence in the infected animals. These studies for the first time demonstrate the interactions between a TLR ligand and iNKT cell activation in visceral leishmaniasis immunotherapeutic. Kala azar (visceral leishmaniasis) is a deadly disease caused by the parasitic protozoa Leishmania donovani. In absence of a suitable vaccine, the incidence of leishmaniasis has increased. The World Health Organization observes that, if the disease is not treated, the fatality rate in developing countries can be as high as 100% within 2 years. Therapy of visceral leishmaniasis can be complicated by toxic side effects, drug resistance, and the need for prolonged treatment regimens. Therefore, improved therapy for leishmaniasis remains desirable. Immunotherapy to selectively induce type 1 immune responses considered essential for resistance to leishmaniasis has shown great promise. CD1d-binding glycolipids stimulate TCR signaling and activation of invariant natural killer T (iNKT) cells. Terminal β-(1–4)-galactose residues in glycoconjugates have been identified as the TLR ligand that induces IFN-γ via TLR signaling. We have used the β-(1–4)-galactose terminal glycosphingophospholipid (GSPL) antigen from L. donovani parasites to treat infected BALB/c mice. We report that immunotherapy with GSPL induced IFN-γ, a type 1 cytokine, through the cooperative action of TLR4 and NKT-cells that contributed to effective control of acute parasite burden in the infected animals.
Collapse
MESH Headings
- Animals
- Antigen Presentation/drug effects
- Antigen Presentation/genetics
- Antigens, CD1d/genetics
- Antigens, CD1d/immunology
- Antigens, CD1d/metabolism
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Antigens, Protozoan/metabolism
- Antigens, Protozoan/pharmacology
- Cricetinae
- Glycosphingolipids/genetics
- Glycosphingolipids/immunology
- Glycosphingolipids/metabolism
- Glycosphingolipids/pharmacology
- Immunotherapy/methods
- Leishmania donovani/immunology
- Leishmania donovani/metabolism
- Leishmaniasis, Visceral/genetics
- Leishmaniasis, Visceral/immunology
- Leishmaniasis, Visceral/therapy
- Mice
- Mice, Inbred BALB C
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/metabolism
- Polysaccharides/genetics
- Polysaccharides/immunology
- Polysaccharides/metabolism
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Toll-Like Receptor 4/genetics
- Toll-Like Receptor 4/immunology
- Toll-Like Receptor 4/metabolism
Collapse
Affiliation(s)
| | | | | | - Tripti De
- Division of Infectious Disease and Immunology, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, Kolkata, India
- * E-mail:
| |
Collapse
|
35
|
Salvador A, Igartua M, Hernández RM, Pedraz JL. Combination of immune stimulating adjuvants with poly(lactide-co-glycolide) microspheres enhances the immune response of vaccines. Vaccine 2012; 30:589-96. [DOI: 10.1016/j.vaccine.2011.11.057] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 11/14/2011] [Accepted: 11/15/2011] [Indexed: 11/30/2022]
|
36
|
Wang D, Xiao PL, Duan HX, Zhou M, Liu J, Li W, Luo KL, Chen JJ, Hu JY. Peptidoglycans promotes human leukemic THP-1 cell apoptosis and differentiation. Asian Pac J Cancer Prev 2012; 13:6409-13. [PMID: 23464467 DOI: 10.7314/apjcp.2012.13.12.6409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The innate immune system coordinates the inflammatory response to pathogens. To do so, its cells must discriminate self from non-self utilizing receptors that identify molecules synthesized exclusively by microbes. Toll- like receptors have a crucial role in the detection of microbial infection in mammals and insects. In mammals, they have evolved to recognize conserved products unique to microbial metabolism. These include lipopolysaccharide (LPS), lipotechoic acids, and peptidoglycans (PGN). We show here that TLRs, including TLR2, are expressed on the THP-1 human leukemia cell line. Activation of TLR2 signaling in THP-1 by PGN induces the synthesis of various soluble factors and proteins including interleukin-1β, interleukin-8 and TNF-α and apoptosis of THP-1 with PGN dose and time dependence. Moreover , in this study we show that PGN induces apoptosis of THP-1 cells in a TNF-α-dependent manner. These findings indicate that TLR2 signaling results in a cascade leading to tumor apoptosis and differentiation, which may suggest new clinical prospects using TLR2 agonists as cytotoxic agents in certain cancers.
Collapse
Affiliation(s)
- Di Wang
- Department of Hematolgy and Oncology, Hunan Provincial People's Hospital, Changsha, China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Godfrey and Rossjohn discuss the varied ways to turn on NKT cells in the context of recent findings. Natural killer T (NKT) cells are CD1d-restricted, lipid antigen–reactive T cells with powerful immunoregulatory potential. The prototypic antigen for NKT cells is a marine sponge–derived glycolipid, α-galactosylceramide (α-GalCer), but this is not normally encountered in the mammalian environment. Thus, there is great interest in the identification of more physiological stimuli for NKT cells, and numerous studies have shown that NKT cells are capable of responding to a range of microbial lipid-based antigens. Two new studies expand our understanding of environmental NKT cell stimuli, with one showing that CD1d-restricted NKT cell antigens are present within common house dust extract (HDE), whereas the other shows that NKT cells can respond to innate stimuli irrespective of the presence of foreign microbial antigens. Collectively, these two investigations indicate that NKT cells are far more likely to encounter foreign antigens, or innate activating signals, than previously recognized, suggesting a more central role for these cells in the immune system.
Collapse
Affiliation(s)
- Dale Ian Godfrey
- Dept. of Microbiology and Immunology, University of Melbourne, Parkville, Victoria 3010, Australia.
| | | |
Collapse
|
38
|
Chiriva-Internati M, Mirandola L, Kast WM, Jenkins MR, Cobos E, Cannon MJ. Understanding the Cross-Talk between Ovarian Tumors and Immune Cells: Mechanisms for Effective Immunotherapies. Int Rev Immunol 2011; 30:71-86. [DOI: 10.3109/08830185.2011.561507] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
39
|
Petersen TR, Sika-Paotonu D, Knight DA, Simkins HMA, Hermans IF. Exploiting the role of endogenous lymphoid-resident dendritic cells in the priming of NKT cells and CD8+ T cells to dendritic cell-based vaccines. PLoS One 2011; 6:e17657. [PMID: 21483862 PMCID: PMC3069042 DOI: 10.1371/journal.pone.0017657] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 02/07/2011] [Indexed: 12/05/2022] Open
Abstract
Transfer of antigen between antigen-presenting cells (APCs) is potentially a physiologically relevant mechanism to spread antigen to cells with specialized stimulatory functions. Here we show that specific CD8+ T cell responses induced in response to intravenous administration of antigen-loaded bone marrow-derived dendritic cells (BM-DCs), were ablated in mice selectively depleted of endogenous lymphoid-resident langerin+ CD8α+ dendritic cells (DCs), suggesting that the antigen is transferred from the injected cells to resident APCs. In contrast, antigen-specific CD4+ T cells were primed predominantly by the injected BM-DCs, with only very weak contribution of resident APCs. Crucially, resident langerin+ CD8α+ DCs only contributed to the priming of CD8+ T cells in the presence of maturation stimuli such as intravenous injection of TLR ligands, or by loading the BM-DCs with the glycolipid α-galactosylceramide (α-GalCer) to recruit the adjuvant activity of activated invariant natural killer-like T (iNKT) cells. In fact, injection of α-GalCer-loaded CD1d−/− BM-DCs resulted in potent iNKT cell activation, suggesting that this glycolipid antigen can also be transferred to resident CD1d+ APCs. While iNKT cell activation per se was independent of langerin+ CD8α+ DCs, some iNKT cell-mediated activities were reduced, notably release of IL-12p70 and transactivation of NK cells. We conclude that both protein and glycolipid antigens can be exchanged between distinct DC species. These data suggest that the efficacy of DC-based vaccination strategies may be improved by the incorporation of a systemic maturation signal aimed to engage resident APCs in CD8+ T cell priming, and α-GalCer may be particularly well suited to this purpose.
Collapse
|
40
|
Vultaggio A, Nencini F, Pratesi S, Maggi L, Guarna A, Annunziato F, Romagnani S, Parronchi P, Maggi E. The TLR7 ligand 9-benzyl-2-butoxy-8-hydroxy adenine inhibits IL-17 response by eliciting IL-10 and IL-10-inducing cytokines. THE JOURNAL OF IMMUNOLOGY 2011; 186:4707-15. [PMID: 21389257 DOI: 10.4049/jimmunol.1002398] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
This study evaluates the ability of a novel TLR7 ligand (9-benzyl-2-butoxy-8-hydroxy adenine, called SA-2) to affect IL-17 response. The SA-2 activity on the expression of IL-17A and IL-17-related molecules was evaluated in acute and chronic models of asthma as well as in in vivo and in vitro α-galactosyl ceramide (α-GalCer)-driven systems. SA-2 prepriming reduced neutrophils in bronchoalveolar lavage fluid and decreased methacoline-induced airway hyperresponsiveness in murine asthma models. These results were associated with the reduction of IL-17A (and type 2 cytokines) as well as of molecules favoring Th17 (and Th2) development in lung tissue. The IL-17A production in response to α-GalCer by spleen mononuclear cells was inhibited in vitro by the presence of SA-2. Reduced IL-17A (as well as IFN-γ and IL-13) serum levels in mice treated with α-GalCer plus SA-2 were also observed. The in vitro results indicated that IL-10 produced by B cells and IL-10-promoting molecules such as IFN-α and IL-27 by dendritic cells are the major player for SA-2-driven IL-17A (and also IFN-γ and IL-13) inhibition. The in vivo experiments with anti-cytokine receptor Abs provided evidence of an early IL-17A inhibition essentially due to IL-10 produced by resident peritoneal cells and of a delayed IL-17A inhibition sustained by IFN-α and IL-27, which in turn drive effector T cells to IL-10 production. These findings suggest that such TLR7 agonist downregulating Th17 (as well as Th2) response has to be considered a valid candidate for novel vaccine formulations in allergy.
Collapse
|
41
|
Moreno M, Mol BM, von Mensdorff-Pouilly S, Verheijen RHM, de Jong EC, von Blomberg BME, van den Eertwegh AJM, Scheper RJ, Bontkes HJ. Differential indirect activation of human invariant natural killer T cells by Toll-like receptor agonists. Immunotherapy 2011; 1:557-70. [PMID: 20635987 DOI: 10.2217/imt.09.30] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AIM Invariant natural killer (iNK) T cells are activated by bacterial glycosphingolipids presented by CD1d on dendritic cells (DCs). Here, it was investigated whether Toll-like receptor (TLR) ligands derived from various microorganisms can either directly or indirectly (through DC activation) activate iNKT cells. MATERIALS & METHODS TLR expression by iNKT cells was examined and the ability of various TLR ligands to activate iNKT cells was evaluated. RESULTS Although human iNKT cells express all TLRs, apart from TLR8, they did not respond directly to TLR ligands. However, iNKT cells became strongly activated when total peripheral blood mononuclear cells were stimulated with TLR2/6, 7/8 and 9 ligands, but not or to a lesser extent with TLR3, 4 and 5 ligands. TLR-stimulated monocyte-derived DCs promoted iNKT cell phenotypic activation and, in turn, these activated iNKT cells further enhanced DC maturation. CONCLUSION TLR agonists may act as strong adjuvants for immunotherapy by promoting combined and reciprocal activation of iNKT cells and DCs.
Collapse
|
42
|
Aldhamen YA, Appledorn DM, Seregin SS, Liu CJJ, Schuldt NJ, Godbehere S, Amalfitano A. Expression of the SLAM family of receptors adapter EAT-2 as a novel strategy for enhancing beneficial immune responses to vaccine antigens. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:722-32. [PMID: 21149608 PMCID: PMC11119279 DOI: 10.4049/jimmunol.1002105] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Recent studies have shown that activation of the signaling lymphocytic activation molecule (SLAM) family of receptors plays an important role in several aspects of immune regulation. However, translation of this knowledge into a useful clinical application has not been undertaken. One important area where SLAM-mediated immune regulation may have keen importance is in the field of vaccinology. Because SLAM signaling plays such a critical role in the innate and adaptive immunity, we endeavored to develop a strategy to improve the efficacy of vaccines by incorporation of proteins known to be important in SLAM-mediated signaling. In this study, we hypothesized that coexpression of the SLAM adapter EWS-FLI1-activated transcript 2 (EAT-2) along with a pathogen-derived Ag would facilitate induction of beneficial innate immune responses, resulting in improved induction of Ag-specific adaptive immune responses. To test this hypothesis, we used rAd5 vector-based vaccines expressing murine EAT-2, or the HIV-1-derived Ag Gag. Compared with appropriate controls, rAd5 vectors expressing EAT-2 facilitated bystander activation of NK, NKT, B, and T cells early after their administration into animals. EAT-2 overexpression also augments the expression of APC (macrophages and dendritic cells) surface markers. Indeed, this multitiered activation of the innate immune system by vaccine-mediated EAT-2 expression enhanced the induction of Ag-specific cellular immune responses. Because both mice and humans express highly conserved EAT-2 adapters, our results suggest that human vaccination strategies that specifically facilitate SLAM signaling may improve vaccine potency when targeting HIV Ags specifically, as well as numerous other vaccine targets in general.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Adaptor Proteins, Signal Transducing
- Adenoviridae Infections/genetics
- Adenoviridae Infections/immunology
- Adenoviridae Infections/therapy
- Adenoviruses, Human/genetics
- Adenoviruses, Human/immunology
- Animals
- Cell Line
- Cells, Cultured
- Genetic Engineering/methods
- Genetic Vectors/administration & dosage
- Genetic Vectors/immunology
- Humans
- Immunity, Cellular/genetics
- Immunity, Innate/genetics
- Intracellular Signaling Peptides and Proteins/administration & dosage
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Multigene Family/immunology
- Signaling Lymphocytic Activation Molecule Associated Protein
- Transcription Factors/administration & dosage
- Transcription Factors/genetics
- Transcription Factors/immunology
- Transcription Factors/physiology
- gag Gene Products, Human Immunodeficiency Virus/genetics
- gag Gene Products, Human Immunodeficiency Virus/immunology
Collapse
Affiliation(s)
- Yasser A Aldhamen
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Girvan RC, Knight DA, O’loughlin CJ, Hayman CM, Hermans IF, Webster GA. MIS416, a non-toxic microparticle adjuvant derived from Propionibacterium acnes comprising immunostimulatory muramyl dipeptide and bacterial DNA promotes cross-priming and Th1 immunity. Vaccine 2011; 29:545-57. [DOI: 10.1016/j.vaccine.2010.10.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 10/14/2010] [Indexed: 10/18/2022]
|
44
|
Caielli S, Conforti-Andreoni C, Di Pietro C, Usuelli V, Badami E, Malosio ML, Falcone M. On/off TLR signaling decides proinflammatory or tolerogenic dendritic cell maturation upon CD1d-mediated interaction with invariant NKT cells. THE JOURNAL OF IMMUNOLOGY 2010; 185:7317-29. [PMID: 21078913 DOI: 10.4049/jimmunol.1000400] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Invariant NKT (iNKT) cells play an effector/adjuvant function during antimicrobial and antitumoral immunity and a regulatory role to induce immune tolerance and prevent autoimmunity. iNKT cells that differentially modulate adaptive immunity do not bear a unique phenotype and/or specific cytokine secretion profile, thus opening questions on how a single T cell subset can exert opposite immunological tasks. In this study, we show that iNKT cells perform their dual roles through a single mechanism of action relying on the cognate interaction with myeloid dendritic cells (DCs) and leading to opposite effects depending on the presence of other maturation stimuli simultaneously acting on DCs. The contact of murine purified iNKT cells with immature autologous DCs directly triggers the tolerogenic maturation of DCs, rendering them able to induce regulatory T cell differentiation and prevent autoimmune diabetes in vivo. Conversely, the interaction of the same purified iNKT cells with DCs, in the presence of simultaneous TLR4 stimulation, significantly enhances proinflammatory DC maturation and IL-12 secretion. The different iNKT cell effects are mediated through distinct mechanisms and activation of different molecular pathways within the DC: CD1d signaling and activation of the ERK1/2 pathway for the tolerogenic action, and CD40-CD40L interaction and NF-κB activation for the adjuvant effect. Our data suggest that the DC decision to undergo proinflammatory or tolerogenic maturation results from the integration of different signals received at the time of iNKT cell contact and could have important therapeutic implications for exploiting iNKT cell adjuvant/regulatory properties in autoimmune diseases, infections, and cancer.
Collapse
Affiliation(s)
- Simone Caielli
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
45
|
Caielli S, Sorini C, Falcone M. The dangerous liaison between iNKT cells and dendritic cells: does it prevent or promote autoimmune diseases? Autoimmunity 2010; 44:11-22. [PMID: 20672910 DOI: 10.3109/08916931003782130] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Invariant natural killer T (iNKT) cells represent an important regulatory T-cell subset that perceives signals of danger and/or cellular distress and modulate the adaptive immune response accordingly. In the presence of pathogens, iNKT cells acquire an adjuvant function that is fundamental to boost anti-microbial and anti-tumor immunity. At the same time, iNKT cells can play a negative regulatory function to maintain peripheral T-cell tolerance toward self-antigens and to prevent autoimmune disease. Both these effects of iNKT cells involve the modulation of the activity of dendritic cells (DCs) through cell-cell interaction. Indeed, iNKT cells can either boost Th1 immunity by enhancing maturation of pro-inflammatory DCs or promote immune tolerance through the maturation of tolerogenic DCs. This dual action of iNKT cells opens questions on the modalities by which a single-cell subset can exert opposite effects on DCs and may even put in question the overall immunosuppressive properties of iNKT cells. This review presents the large body of evidence that shows the ability of iNKT cells to negatively regulate autoimmunity and to prevent autoimmune diseases including multiple sclerosis, type 1 diabetes, rheumatoid arthritis, and systemic lupus erythematosus. In addition, an update is provided on the mechanisms of iNKT-DCs interactions and how this can result in inflammatory or tolerogenic responses.
Collapse
Affiliation(s)
- Simone Caielli
- Experimental Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | | | | |
Collapse
|
46
|
Noda K, Kodama S, Umemoto S, Abe N, Hirano T, Suzuki M. Nasal vaccination with P6 outer membrane protein and alpha-galactosylceramide induces nontypeable Haemophilus influenzae-specific protective immunity associated with NKT cell activation and dendritic cell expansion in nasopharynx. Vaccine 2010; 28:5068-74. [PMID: 20478344 DOI: 10.1016/j.vaccine.2010.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Revised: 04/29/2010] [Accepted: 05/03/2010] [Indexed: 01/10/2023]
Abstract
The efficacy of alpha-galactosylceramide (alpha-GalCer) as a mucosal adjuvant was examined. Mice were immunized intranasally with nontypeable Haemophilus influenzae (NTHi) P6 protein and alpha-GalCer. P6-specific antibody responses in the form of P6-specific IgA in nasal washes and serum IgG titers were significantly elevated. Splenic CD4(+) T cells expressed P6-specific Th1 and Th2 cytokine mRNA. In addition, NTHi was quantified in nasal washes following NTHi challenges, and the clearance of NTHi from the nasopharynx was also enhanced. These results indicate that alpha-GalCer might be an effective mucosal adjuvant.
Collapse
Affiliation(s)
- Kenji Noda
- Department of Otolaryngology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hazama-machi, Yufu, Oita 879-5593, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Xiong G, Husseiny MI, Song L, Erdreich-Epstein A, Shackleford GM, Seeger RC, Jäckel D, Hensel M, Metelitsa LS. Novel cancer vaccine based on genes of Salmonella pathogenicity island 2. Int J Cancer 2010; 126:2622-34. [PMID: 19824039 DOI: 10.1002/ijc.24957] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although tumors express potentially immunogenic tumor-associated antigens (TAAs), cancer vaccines often fail because of inadequate antigen delivery and/or insufficient activation of innate immunity. Engineering nonpathogenic bacterial vectors to deliver TAAs of choice may provide an efficient way of presenting TAAs in an immunogenic form. In this study, we used genes of Salmonella pathogenicity island 2 (SPI2) to construct a novel cancer vaccine in which a TAA, survivin, was fused to SseF effector protein and placed under control of SsrB, the central regulator of SPI2 gene expression. This construct uses the type III secretion system (T3SS) of Salmonella and allows preferential delivery of tumor antigen into the cytosol of antigen-presenting cells for optimal immunogenicity. In a screen of a panel of attenuated strains of Salmonella, we found that a double attenuated strain of Salmonella typhimurium, MvP728 (purD/htrA), was not toxic to mice and effectively expressed and translocated survivin protein inside the cytosol of murine macrophages. We also found that a ligand for CD1d-reactive natural killer T (NKT) cells, alpha-glucuronosylceramide (GSL1), enhanced MvP728-induced interleukin-12 production in human dendritic cells and that in vivo coadministration of a NKT ligand with MvP728-Llo or MvP728-survivin enhanced effector-memory cytotoxic T lymphocyte (CTL) responses. Furthermore, combined use of MvP728-survivin with GSL1 produced antitumor activity in mouse models of CT26 colon carcinoma and orthotopic DBT glioblastoma. Therefore, the use of TAA delivery via SPI-2-regulated T3SS of Salmonella and NKT ligands as adjuvants may provide a foundation for new cancer vaccines.
Collapse
Affiliation(s)
- Guosheng Xiong
- Division of Hematology-Oncology, Department of Pediatrics, University of Southern California Keck School of Medicine and The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Potent anti-tumor responses to immunization with dendritic cells loaded with tumor tissue and an NKT cell ligand. Immunol Cell Biol 2010; 88:596-604. [PMID: 20142835 DOI: 10.1038/icb.2010.9] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy is well tolerated and specific, but its efficacy remains variable. To enhance anti-tumor CD8(+) T-cell responses induced by immunization with antigen-loaded dendritic cells (DCs), we explored the impact of eliciting a potent source of T-cell help from activated invariant natural killer (NK)-like T cells (iNKT cells) using the specific glycolipid ligand alpha-galactosylceramide (alpha-GalCer). As cytokines released by iNKT cells may drive proliferation of CD4(+)CD25(+) regulatory T cells (Tregs), we assessed this immunization strategy in animals treated with anti-CD25 antibody to inactivate Treg function. Combining DC immunization with iNKT cell activation was found to significantly enhance anti-tumor activity, which was improved further by the prior inactivation of Tregs. The improved anti-tumor activity with Treg inactivation was associated with a prolonged proliferative burst of responding CD8(+) T cells. We could find no evidence that inclusion of alpha-GalCer in the vaccine enhanced Treg numbers, or that the 'helper' function of iNKT cells was improved in the absence of Treg activity. Rather, the two activities appeared to act independently to improve the tumor-specific T-cell response. Inactivating regulatory T cells and eliciting iNKT cell activation are therefore two useful strategies that can be used in combination to improve anti-tumor immunization with antigen-loaded DCs.
Collapse
|
49
|
Joyee AG, Uzonna J, Yang X. Invariant NKT Cells Preferentially Modulate the Function of CD8α+ Dendritic Cell Subset in Inducing Type 1 Immunity against Infection. THE JOURNAL OF IMMUNOLOGY 2010; 184:2095-106. [DOI: 10.4049/jimmunol.0901348] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
50
|
Bhogal RH, Afford SC. Immune Cell Communication and Signaling Systems in Liver Disease. SIGNALING PATHWAYS IN LIVER DISEASES 2010:117-146. [DOI: 10.1007/978-3-642-00150-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|