1
|
Palacios PA, Santibañez Á, Aguirre-Muñoz F, Gutiérrez-Vera C, Niño de Zepeda-Carrizo V, Góngora-Pimentel M, Müller M, Cáceres M, Kalergis AM, Carreño LJ. Can invariant Natural Killer T cells drive B cell fate? a look at the humoral response. Front Immunol 2025; 16:1505883. [PMID: 40040714 PMCID: PMC11876049 DOI: 10.3389/fimmu.2025.1505883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/21/2025] [Indexed: 03/06/2025] Open
Abstract
Invariant Natural Killer T (NKT) cells represent a unique subset of innate-like T cells that express both NK cell and T cell receptors. These cells are rapidly activated by glycolipid antigens presented via CD1d molecules on antigen-presenting cells (APCs), including B cells, dendritic cells (DCs), and macrophages, or through cytokine-dependent mechanisms. Their ability to produce a wide range of cytokines and express costimulatory molecules underscores their critical role in bridging innate and adaptive immunity. B cells, traditionally recognized for their role in antibody production, also act as potent APCs due to their high expression of CD1d, enabling direct interactions with iNKT cells. This interaction has significant implications for humoral immunity, influencing B cell activation, class-switch recombination (CSR), germinal center formation, and memory B cell differentiation, thus expanding the conventional paradigm of T cell-B cell interactions. While the influence of iNKT cells on B cell biology and humoral responses is well-supported, many aspects of their interaction remain unresolved. Key questions include the roles of different iNKT cell subsets, the diversity of APCs, the spatiotemporal dynamics of these interactions, especially during early activation, and the potential for distinct glycolipid ligands to modulate immune outcomes. Understanding these factors could provide valuable insights into how iNKT cells regulate B cell-mediated immunity and offer opportunities to harness these interactions in immunotherapeutic applications, such as vaccine development. In this review, we examine these unresolved aspects and propose a novel perspective on the regulatory potential of iNKT cells in humoral immunity, emphasizing their promise as a target for innovative vaccine strategies.
Collapse
Affiliation(s)
- Pablo A. Palacios
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Álvaro Santibañez
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Fernanda Aguirre-Muñoz
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Cristián Gutiérrez-Vera
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Valentina Niño de Zepeda-Carrizo
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Martín Góngora-Pimentel
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marioly Müller
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mónica Cáceres
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leandro J. Carreño
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
2
|
O’Hara MP, Yanamandra AV, Sastry KJ. Immunity from NK Cell Subsets Is Important for Vaccine-Mediated Protection in HPV+ Cancers. Vaccines (Basel) 2024; 12:206. [PMID: 38400189 PMCID: PMC10892709 DOI: 10.3390/vaccines12020206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
High-risk human papillomaviruses (HPVs) are associated with genital and oral cancers, and the incidence of HPV+ head and neck squamous cell cancers is fast increasing in the USA and worldwide. Survival rates for patients with locally advanced disease are poor after standard-of-care chemoradiation treatment. Identifying the antitumor host immune mediators important for treatment response and designing strategies to promote them are essential. We reported earlier that in a syngeneic immunocompetent preclinical HPV tumor mouse model, intranasal immunization with an HPV peptide therapeutic vaccine containing the combination of aGalCer and CpG-ODN adjuvants (TVAC) promoted clearance of HPV vaginal tumors via induction of a strong cytotoxic T cell response. However, TVAC was insufficient in the clearance of HPV oral tumors. To overcome this deficiency, we tested substituting aGalCer with a clinically relevant adjuvant QS21 (TVQC) and observed sustained, complete regression of over 70% of oral and 80% of vaginal HPV tumors. The TVQC-mediated protection in the oral tumor model correlated with not only strong total and HPV-antigen-specific CD8 T cells, but also natural killer dendritic cells (NKDCs), a novel subset of NK cells expressing the DC marker CD11c. Notably, we observed induction of significantly higher overall innate NK effector responses by TVQC relative to TVAC. Furthermore, in mice treated with TVQC, the frequencies of total and functional CD11c+ NK cell populations were significantly higher than the CD11c- subset, highlighting the importance of the contributions of NKDCs to the vaccine response. These results emphasize the importance of NK-mediated innate immune effector responses in total antitumor immunity to treat HPV+ cancers.
Collapse
Affiliation(s)
- Madison P. O’Hara
- Department of Thoracic Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.P.O.); (A.V.Y.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ananta V. Yanamandra
- Department of Thoracic Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.P.O.); (A.V.Y.)
| | - K. Jagannadha Sastry
- Department of Thoracic Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.P.O.); (A.V.Y.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
3
|
Zhao W, Li M, Song S, Zhi Y, Huan C, Lv G. The role of natural killer T cells in liver transplantation. Front Cell Dev Biol 2024; 11:1274361. [PMID: 38250325 PMCID: PMC10796773 DOI: 10.3389/fcell.2023.1274361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024] Open
Abstract
Natural killer T cells (NKTs) are innate-like lymphocytes that are abundant in the liver and participate in liver immunity. NKT cells express both NK cell and T cell markers, modulate innate and adaptive immune responses. Type I and Type II NKT cells are classified according to the TCR usage, while they recognize lipid antigen in a non-classical major histocompatibility (MHC) molecule CD1d-restricted manner. Once activated, NKT cells can quickly produce cytokines and chemokines to negatively or positively regulate the immune responses, depending on the different NKT subsets. In liver transplantation (LTx), the immune reactions in a series of processes determine the recipients' long-term survival, including ischemia-reperfusion injury, alloresponse, and post-transplant infection. This review provides insight into the research on NKT cells subpopulations in LTx immunity during different processes, and discusses the shortcomings of the current research on NKT cells. Additionally, the CD56-expressing T cells are recognized as a NK-like T cell population, they were also discussed during these processes.
Collapse
Affiliation(s)
- Wenchao Zhao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Mingqian Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Shifei Song
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yao Zhi
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Chen Huan
- Center of Infectious Diseases and Pathogen Biology, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
4
|
Fujii SI, Shimizu K. NKT-Licensed In Vivo Dendritic Cell-Based Immunotherapy as Cellular Immunodrugs for Cancer Treatment. Crit Rev Oncog 2024; 29:45-61. [PMID: 38421713 DOI: 10.1615/critrevoncog.2023048735] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
With the advent of new therapies, immunotherapy has gained attention as a critical modality. After the discovery of the natural killer T (NKT) cells ligand, ex vivo cultured dendritic cells (DCs) loaded with NKT ligand (especially α-galactosylceramide (α-GalCer) (DC/Gal) or ex vivo expanded NKT transfer studies were clinically examined in several institutes. To prevent tumoral immune escape, the link between innate and adaptive immunity, in situ selective targeting of DCs has been attempted; however, protocol optimization was required. As a type of DC targeting therapy that combines the benefits of invariant natural killer T (iNKT) cells, we established an all-in-one, off-the-shelf drug, named the artificial adjuvant vector cell (aAVC), which consists of the tumor antigen and the CD1d-iNKT ligand complex. Here, to our knowledge, we first demonstrate the DC/GalCer therapy and NKT transfer therapy. Next, we introduce and discuss the use of aAVC therapy not only for efficient innate and adaptive immunity induction using fully matured DC in situ but also the characterization necessary for locally reprogramming the tumor microenvironment and systemically inducing long-term memory in T cells. We also discuss how the immune network mechanism is controlled by DCs. Next, we performed the first human clinical trial using WT1 antigen-expressing aAVC against relapse and refractory acute myelogenous leukemia. Thus, we highlight the challenges of using aAVCs as prodrugs for actively energizing DCs in vivo, underpinning immunological networks, and developing strategies for providing maximal benefits for patients.
Collapse
Affiliation(s)
- Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), and RIKEN Program for Drug Discovery and Medical Technology Platforms, Yokohama, Kanagawa, Japan
| | - Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), and RIKEN Program for Drug Discovery and Medical Technology Platforms, Yokohama, Kanagawa, Japan
| |
Collapse
|
5
|
Alhamawi RM, Aloufi N, Alamri AF, Altubayli FA, Alsairi RT, Alhamad RA, Alharbi SM, Ankhli ZA, Eid HMA, Almutawif YA. Prognostic impact of invariant natural killer T cells in solid and hematological tumors; systematic review and meta-analysis. Cancer Biomark 2024; 41:155-164. [PMID: 39302356 PMCID: PMC11492061 DOI: 10.3233/cbm-240069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/18/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Invariant natural killer T (iNKT) cells are an immune subset that purportedly link the adaptive and the innate arms of the immune system. Importantly, iNKT cells contribute to anti-cancer immunity in different types of hematological and solid malignancies by secreting pro-inflammatory cytokines. Therefore, using such cells in treating different type of tumors would be an ideal candidate for cancer immunotherapy. OBJECTIVE To assess the prognostic effect of iNKT cells across different types of solid and hematological tumors. METHODS In systematic review and meta-analysis, articles assessed the prognostic effect of iNKT cells were systemically searched using the scientific databases including Google Scholar, ScienceDirect, PubMed, Cochrane Central, and Scopus. RESULTS Strikingly, the analysis showed the positive impact of intratumoral or circulating iNKT cells on the survival rate in patients with all studied tumors with overall effect of a pooled hazard ratio of 0.89 (95% CI 0.81 to 0.98; p= 0.01). A highly statistical heterogeneity was noted between studied tumor with I2 = 87%; p= 0.00001. CONCLUSIONS Taken together, this study would present a new insight into the impact of iNKT cells correlate with caner patients' survival rate and how such cells would be used as a therapeutic target in these patients.
Collapse
Affiliation(s)
- Renad M. Alhamawi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Noof Aloufi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Abeer F. Alamri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Fatima A. Altubayli
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Raghad T. Alsairi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Reem A. Alhamad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Shouq M. Alharbi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Zainab A. Ankhli
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Hamza M. A. Eid
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Yahya A. Almutawif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| |
Collapse
|
6
|
Heuser-Loy C, Baumgart AK, Hackstein CP, Courrèges CJF, Philipp MS, Thaiss CA, Holland T, Evaristo C, Garbi N, Kurts C. Conditional NKT Cell Depletion in Mice Reveals a Negative Feedback Loop That Regulates CTL Cross-Priming. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:35-42. [PMID: 38019126 DOI: 10.4049/jimmunol.2300662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/29/2023] [Indexed: 11/30/2023]
Abstract
NKT cells are unconventional T cells whose biological role is incompletely understood. Similar to TH cells, activated NKT cells can cause dendritic cell (DC) maturation, which is required for effective CTL responses. However, it is unclear whether and how NKT cells affect CTLs downstream of the DC maturation phase. This is partially due to the lack of techniques to conditionally deplete NKT cells in vivo. To overcome this problem, we have developed two approaches for this purpose in mice: the first is based on mixed bone marrow chimeras where Jα18 knockout and depletable CD90 congenic bone marrow is combined, and the second used PLZFCre × iDTR bone marrow chimeras, which target innate-like T cells. Using these tools, we found that NKT cell depletion at 20 h, that is, after initial DC activation, did not render CTLs helpless, as CD40L signaling by non-NKT cells sufficed. Instead, NKT cell depletion even augmented CD8 T cell expansion and cytotoxicity by mechanisms distinct from reduced STAT6 signaling. These findings revealed a negative feedback loop by which NKT cells control CTL cross-priming downstream of DC maturation. The techniques described in this study expand the toolbox to study NKT cells and other unconventional T cell subsets in vivo and uncovered a hidden immunoregulatory mechanism.
Collapse
Affiliation(s)
- Christoph Heuser-Loy
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Ann-Kathrin Baumgart
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Carl-Philipp Hackstein
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Christina J F Courrèges
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Marie-Sophie Philipp
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Christoph A Thaiss
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Tristan Holland
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - César Evaristo
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Natalio Garbi
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Christian Kurts
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
- The Peter Doherty Institute of Infection and Immunology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
7
|
Burn OK, Dasyam N, Hermans IF. Recruiting Natural Killer T Cells to Improve Vaccination: Lessons from Preclinical and Clinical Studies. Crit Rev Oncog 2024; 29:31-43. [PMID: 38421712 DOI: 10.1615/critrevoncog.2023049407] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The capacity of type I natural killer T (NKT) cells to provide stimulatory signals to antigen-presenting cells has prompted preclinical research into the use of agonists as immune adjuvants, with much of this work focussed on stimulating T cell responses to cancer. In attempting to evaluate this approach in the clinic, our recent dendritic-cell based study failed to show an advantage to adding an agonist to the vaccine. Here we present potential limitations of the study, and suggest why other simpler strategies may be more effective. These include strategies to target antigen-presenting cells in the host, either through promoting efficient transfer from injected cell lines, facilitating uptake of antigen and agonist as injected conjugates, or encapsulating the components into injected nanovectors. While the vaccine landscape has changed with the rapid uptake of mRNA vaccines, we suggest that there is still a role for recruiting NKT cells in altering T cell differentiation programmes, notably the induction of resident memory T cells.
Collapse
Affiliation(s)
- Olivia K Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
8
|
Lin Q, Wang T, Zuo X, Ni H, Zhong J, Zhan L, Cheng H, Huang Y, Ding X, Yu H, Nie H. Anti-CD1d treatment suppresses immunogenic maturation of lung dendritic cells dependent on lung invariant natural killer T cells in asthmatic mice. Int Immunopharmacol 2023; 124:110921. [PMID: 37725846 DOI: 10.1016/j.intimp.2023.110921] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/21/2023]
Abstract
Our previous findings show that invariant natural killer T (iNKT)cells can promote immunogenic maturation of lung dendritic cells (LDCs) to enhance Th2 cell responses in asthma. It has been accepted that recognition of glycolipid antigens presented by CD1d molecules by the T cell receptors of iNKT cells leads to iNKT cell activation. Therefore, we examine the immunoregulatory influences of anti-CD1d treatment on Th2 cell response and immunogenic maturation of LDCs and subsequently explored whether these influences were dependent on lung iNKT cells in asthmatic mice. We discoveredthat in wild-type mice sensitized and challenged with house dust mite or ovalbumin (OVA), anti-CD1d treatment inhibited Th2 cell response and immunogenic maturation of LDCs. LDCs from asthmatic mice with anti-CD1d treatment had a markedly decreased influence on Th2 cell responses in vivo and in vitro. Furthermore, anti-CD1d treatment reduced the abundance and activation of lung iNKT cells in asthmatic mice. Moreover, in asthmatic iNKT cell-deficient Jα18-/- mice, anti-CD1d treatment did not influence Th2 cell responses and immunogenic maturation of LDCs. Meanwhile, the quantity of CD40L+ iNKT cells in asthmatic mice was significant decreased by anti-CD1d treatment. Finally, the inhibition of anti-CD1d treatment on LDC immunogenic maturation and Th2 cell responses in asthmatic mice was reversed by anti-CD40 treatment. Our data suggest that anti-CD1d treatment can suppress Th2 cell responses through inhibiting immunogenic maturation of LDCs dependent on lung iNKT cells, which couldbe partially related to the downregulation of CD40L expression on lung iNKT cells in asthmatic mice.
Collapse
Affiliation(s)
- Qibin Lin
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Tong Wang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Xiaoshu Zuo
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Haiyang Ni
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Jieying Zhong
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Liying Zhan
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Hong Cheng
- Department of Parmacy, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Yi Huang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Xuhong Ding
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Hongying Yu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Hanxiang Nie
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China.
| |
Collapse
|
9
|
Gonzatti MB, Freire BM, Antunes MM, de Menezes GB, Talbot J, Peron JPS, Basso AS, Keller AC. Targeting adrenergic receptors to mitigate invariant natural killer T cells-induced acute liver injury. iScience 2023; 26:107947. [PMID: 37841583 PMCID: PMC10568435 DOI: 10.1016/j.isci.2023.107947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/07/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023] Open
Abstract
Invariant Natural Killer T (iNKT) cell activation by α-galactosylceramide (αGC) potentiates cytotoxic immune responses against tumors. However, αGC-induced liver injury is a limiting factor for iNKT-based immunotherapy. Although adrenergic receptor stimulation is an important immunosuppressive signal that curbs tissue damage induced by inflammation, its effect on the antitumor activity of invariant Natural Killer T (iNKT) cells remains unclear. We use mouse models and pharmacological tools to show that the stimulation of the sympathetic nervous system (SNS) inhibits αGC-induced liver injury without impairing iNKT cells' antitumoral functions. Mechanistically, SNS stimulation prevents the collateral effect of TNF-α production by iNKT cells and neutrophil accumulation in hepatic parenchyma. Our results suggest that the modulation of the adrenergic signaling can be a complementary approach to αGC-based immunotherapy to mitigate iNKT-induced liver injury without compromising its antitumoral activity.
Collapse
Affiliation(s)
- Michelangelo Bauwelz Gonzatti
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), Rua Botucatu, 862, 4th floor, São Paulo 04023-062, Brazil
| | - Beatriz Marton Freire
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), Rua Botucatu, 862, 4th floor, São Paulo 04023-062, Brazil
| | - Maísa Mota Antunes
- Department of Morphology, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Minas Gerais 31270-910, Brazil
| | - Gustavo Batista de Menezes
- Department of Morphology, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Minas Gerais 31270-910, Brazil
| | - Jhimmy Talbot
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA 98109-1024, USA
| | - Jean Pierre Schatzmann Peron
- Department of Immunology-ICB IV, University of São Paulo, Av. Prof. Lineu Prestes, 1730, São Paulo 05508-900, Brazil
| | - Alexandre Salgado Basso
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), Rua Botucatu, 862, 4th floor, São Paulo 04023-062, Brazil
| | - Alexandre Castro Keller
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), Rua Botucatu, 862, 4th floor, São Paulo 04023-062, Brazil
| |
Collapse
|
10
|
Tsuji M, Nair MS, Masuda K, Castagna C, Chong Z, Darling TL, Seehra K, Hwang Y, Ribeiro ÁL, Ferreira GM, Corredor L, Coelho-Dos-Reis JGA, Tsuji Y, Mori M, Boon ACM, Diamond MS, Huang Y, Ho DD. An immunostimulatory glycolipid that blocks SARS-CoV-2, RSV, and influenza infections in vivo. Nat Commun 2023; 14:3959. [PMID: 37402814 DOI: 10.1038/s41467-023-39738-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023] Open
Abstract
Prophylactic vaccines for SARS-CoV-2 have lowered the incidence of severe COVID-19, but emergence of viral variants that are antigenically distinct from the vaccine strains are of concern and additional, broadly acting preventive approaches are desirable. Here, we report on a glycolipid termed 7DW8-5 that exploits the host innate immune system to enable rapid control of viral infections in vivo. This glycolipid binds to CD1d on antigen-presenting cells and thereby stimulates NKT cells to release a cascade of cytokines and chemokines. The intranasal administration of 7DW8-5 prior to virus exposure significantly blocked infection by three different authentic variants of SARS-CoV-2, as well as by respiratory syncytial virus and influenza virus, in mice or hamsters. We also found that this protective antiviral effect is both host-directed and mechanism-specific, requiring both the CD1d molecule and interferon-[Formula: see text]. A chemical compound like 7DW8-5 that is easy to administer and cheap to manufacture may be useful not only in slowing the spread of COVID-19 but also in responding to future pandemics long before vaccines or drugs are developed.
Collapse
Affiliation(s)
- Moriya Tsuji
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Manoj S Nair
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Kazuya Masuda
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Candace Castagna
- Institute of Comparative Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Zhenlu Chong
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tamarand L Darling
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kuljeet Seehra
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Youngmin Hwang
- Columbia Center for Human Development, Pulmonary Allergy & Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Ágata Lopes Ribeiro
- Basic and Applied Virology Laboratory, Department of Microbiology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Geovane Marques Ferreira
- Basic and Applied Virology Laboratory, Department of Microbiology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Laura Corredor
- Institute of Comparative Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | | | - Yukiko Tsuji
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Munemasa Mori
- Columbia Center for Human Development, Pulmonary Allergy & Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Adrianus C M Boon
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA.
| |
Collapse
|
11
|
Dasyam N, Sharples KJ, Barrow C, Huang Y, Bauer E, Mester B, Wood CE, Authier-Hall A, Dzhelali M, Ostapowicz T, Kumar R, Lowe J, Maxwell A, Burn OK, Williams GM, Carley SE, Caygill G, Jones J, Chan STS, Hinder VA, Macapagal J, McCusker M, Weinkove R, Brimble MA, Painter GF, Findlay MP, Dunbar PR, Gasser O, Hermans IF. A randomised controlled trial of long NY-ESO-1 peptide-pulsed autologous dendritic cells with or without alpha-galactosylceramide in high-risk melanoma. Cancer Immunol Immunother 2023; 72:2267-2282. [PMID: 36881133 PMCID: PMC10264280 DOI: 10.1007/s00262-023-03400-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 02/06/2023] [Indexed: 03/08/2023]
Abstract
AIM We have previously reported that polyfunctional T cell responses can be induced to the cancer testis antigen NY-ESO-1 in melanoma patients injected with mature autologous monocyte-derived dendritic cells (DCs) loaded with long NY-ESO-1-derived peptides together with α-galactosylceramide (α-GalCer), an agonist for type 1 Natural Killer T (NKT) cells. OBJECTIVE To assess whether inclusion of α-GalCer in autologous NY-ESO-1 long peptide-pulsed DC vaccines (DCV + α-GalCer) improves T cell responses when compared to peptide-pulsed DC vaccines without α-GalCer (DCV). DESIGN, SETTING AND PARTICIPANTS Single-centre blinded randomised controlled trial in patients ≥ 18 years old with histologically confirmed, fully resected stage II-IV malignant cutaneous melanoma, conducted between July 2015 and June 2018 at the Wellington Blood and Cancer Centre of the Capital and Coast District Health Board. INTERVENTIONS Stage I. Patients were randomised to two cycles of DCV or DCV + α-GalCer (intravenous dose of 10 × 106 cells, interval of 28 days). Stage II. Patients assigned to DCV + α-GalCer were randomised to two further cycles of DCV + α-GalCer or observation, while patients initially assigned to DCV crossed over to two cycles of DCV + α-GalCer. OUTCOME MEASURES Primary: Area under the curve (AUC) of mean NY-ESO-1-specific T cell count detected by ex vivo IFN-γ ELISpot in pre- and post-treatment blood samples, compared between treatment arms at Stage I. Secondary: Proportion of responders in each arm at Stage I; NKT cell count in each arm at Stage I; serum cytokine levels at Stage I; adverse events Stage I; T cell count for DCV + α-GalCer versus observation at Stage II, T cell count before versus after cross-over. RESULTS Thirty-eight patients gave written informed consent; 5 were excluded before randomisation due to progressive disease or incomplete leukapheresis, 17 were assigned to DCV, and 16 to DCV + α-GalCer. The vaccines were well tolerated and associated with increases in mean total T cell count, predominantly CD4+ T cells, but the difference between the treatment arms was not statistically significant (difference - 6.85, 95% confidence interval, - 21.65 to 7.92; P = 0.36). No significant improvements in T cell response were associated with DCV + α-GalCer with increased dosing, or in the cross-over. However, the NKT cell response to α-GalCer-loaded vaccines was limited compared to previous studies, with mean circulating NKT cell levels not significantly increased in the DCV + α-GalCer arm and no significant differences in cytokine response between the treatment arms. CONCLUSIONS A high population coverage of NY-ESO-1-specific T cell responses was achieved with a good safety profile, but we failed to demonstrate that loading with α-GalCer provided an additional advantage to the T cell response with this cellular vaccine design. CLINICAL TRIAL REGISTRATION ACTRN12612001101875. Funded by the Health Research Council of New Zealand.
Collapse
Affiliation(s)
- Nathaniel Dasyam
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Katrina J Sharples
- Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
- Cancer Trials New Zealand, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Catherine Barrow
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Ying Huang
- Cancer Trials New Zealand, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Evelyn Bauer
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Brigitta Mester
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Catherine E Wood
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Astrid Authier-Hall
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Marina Dzhelali
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Tess Ostapowicz
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Rajiv Kumar
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Jessica Lowe
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Alice Maxwell
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Olivia K Burn
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Geoffrey M Williams
- School of Chemical Sciences, University of Auckland, PO Box 92019, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Private Bag 92019, Auckland, 1142, New Zealand
| | - Sarah E Carley
- School of Chemical Sciences, University of Auckland, PO Box 92019, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Private Bag 92019, Auckland, 1142, New Zealand
| | | | - Jeremy Jones
- GlycoSyn, PO Box 31 310, Lower Hutt, 5040, New Zealand
| | - Susanna T S Chan
- The Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Lower Hutt, 5046, New Zealand
| | - Victoria A Hinder
- Cancer Trials New Zealand, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Jerome Macapagal
- Cancer Trials New Zealand, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Monica McCusker
- Cancer Trials New Zealand, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Robert Weinkove
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Margaret A Brimble
- School of Chemical Sciences, University of Auckland, PO Box 92019, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Private Bag 92019, Auckland, 1142, New Zealand
| | - Gavin F Painter
- The Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Lower Hutt, 5046, New Zealand
| | - Michael P Findlay
- Cancer Trials New Zealand, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - P Rod Dunbar
- Maurice Wilkins Centre for Molecular Biodiscovery, Private Bag 92019, Auckland, 1142, New Zealand
- School of Biological Sciences, University of Auckland, PO Box 92019, Auckland, New Zealand
| | - Olivier Gasser
- 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.
- Maurice Wilkins Centre for Molecular Biodiscovery, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
12
|
Hadiloo K, Tahmasebi S, Esmaeilzadeh A. CAR-NKT cell therapy: a new promising paradigm of cancer immunotherapy. Cancer Cell Int 2023; 23:86. [PMID: 37158883 PMCID: PMC10165596 DOI: 10.1186/s12935-023-02923-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
Today, cancer treatment is one of the fundamental problems facing clinicians and researchers worldwide. Efforts to find an excellent way to treat this illness continue, and new therapeutic strategies are developed quickly. Adoptive cell therapy (ACT) is a practical approach that has been emerged to improve clinical outcomes in cancer patients. In the ACT, one of the best ways to arm the immune cells against tumors is by employing chimeric antigen receptors (CARs) via genetic engineering. CAR equips cells to target specific antigens on tumor cells and selectively eradicate them. Researchers have achieved promising preclinical and clinical outcomes with different cells by using CARs. One of the potent immune cells that seems to be a good candidate for CAR-immune cell therapy is the Natural Killer-T (NKT) cell. NKT cells have multiple features that make them potent cells against tumors and would be a powerful replacement for T cells and natural killer (NK) cells. NKT cells are cytotoxic immune cells with various capabilities and no notable side effects on normal cells. The current study aimed to comprehensively provide the latest advances in CAR-NKT cell therapy for cancers.
Collapse
Affiliation(s)
- Kaveh Hadiloo
- Student Research Committee, Department of immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of immunology, School of Medicine, Shahid beheshti University of Medical Sciences, Tehran, Iran.
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran.
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
13
|
Yi Q, Wang J, Liu T, Yao Y, Loveless I, Subedi K, Toor J, Adrianto I, Xiao H, Chen B, Crawford HC, Fang D, Zhou L, Mi QS. scRNA-Seq and imaging mass cytometry analyses unveil iNKT cells-mediated anti-tumor immunity in pancreatic cancer liver metastasis. Cancer Lett 2023; 561:216149. [PMID: 36990268 PMCID: PMC11737350 DOI: 10.1016/j.canlet.2023.216149] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/18/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023]
Abstract
Invariant natural killer T (iNKT) cells are innate-like T cells that are abundant in liver sinusoids and play a critical role in tumor immunity. However, the role of iNKT cells in pancreatic cancer liver metastasis (PCLM) has not been fully explored. In this study, we employed a hemi-spleen pancreatic tumor cell injection mouse model of PCLM, a model that closely mimics clinical conditions in humans, to explore the role of iNKT cells in PCLM. Activation of iNKT cells with α-galactosylceramide (αGC) markedly increased immune cell infiltration and suppressed PCLM progression. Via single cell RNA sequencing (scRNA-seq) we profiled over 30,000 immune cells from normal liver and PCLM with or without αGC treatment and were able to characterize the global changes of the immune cells in the tumor microenvironment upon αGC treatment, identifying a total of 12 subpopulations. Upon treatment with αGC, scRNA-Seq and flow cytometry analyses revealed increased cytotoxic activity of iNKT/NK cells and skewing CD4 T cells towards a cytotoxic Th1 profile and CD8 T cells towards a cytotoxic profile, characterized by higher proliferation and reduced exhaustion marker PD1 expression. Moreover, αGC treatment excluded tumor associated macrophages. Lastly, imaging mass cytometry analysis uncovered the reduced epithelial to mesenchymal transition related markers and increased active CD4 and CD8 T cells in PCLM with αGC treatment. Overall, our findings uncover the protective function of activated iNKT cells in pancreatic cancer liver metastasis through increased NK and T cell immunity and decreased tumor associated macrophages.
Collapse
Affiliation(s)
- Qijun Yi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Jie Wang
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Tingting Liu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Yi Yao
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Ian Loveless
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA; Center for Bioinformatics, Department of Public Health Sciences, Henry Ford Health, Detroit, MI, 48202, USA
| | - Kalpana Subedi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Jugmohit Toor
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Indra Adrianto
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA; Center for Bioinformatics, Department of Public Health Sciences, Henry Ford Health, Detroit, MI, 48202, USA; Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Hua Xiao
- Department of Physiology, College of Natural Science, Michigan State University, East Lansing, MI, 48824, USA
| | - Bin Chen
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, USA
| | - Howard C Crawford
- Henry Ford Pancreatic Cancer Center, Department of Surgery, Henry Ford Health, Detroit, MI, 48202, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Li Zhou
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA; Center for Bioinformatics, Department of Public Health Sciences, Henry Ford Health, Detroit, MI, 48202, USA; Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA; Department of Internal Medicine, Henry Ford Health, Detroit, MI, 48202, USA.
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health, Detroit, MI, 48202, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA; Center for Bioinformatics, Department of Public Health Sciences, Henry Ford Health, Detroit, MI, 48202, USA; Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA; Department of Internal Medicine, Henry Ford Health, Detroit, MI, 48202, USA.
| |
Collapse
|
14
|
Pankhurst TE, Buick KH, Lange JL, Marshall AJ, Button KR, Palmer OR, Farrand KJ, Montgomerie I, Bird TW, Mason NC, Kuang J, Compton BJ, Comoletti D, Salio M, Cerundolo V, Quiñones-Mateu ME, Painter GF, Hermans IF, Connor LM. MAIT cells activate dendritic cells to promote T FH cell differentiation and induce humoral immunity. Cell Rep 2023; 42:112310. [PMID: 36989114 PMCID: PMC10045373 DOI: 10.1016/j.celrep.2023.112310] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/02/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
Protective immune responses against respiratory pathogens, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza virus, are initiated by the mucosal immune system. However, most licensed vaccines are administered parenterally and are largely ineffective at inducing mucosal immunity. The development of safe and effective mucosal vaccines has been hampered by the lack of a suitable mucosal adjuvant. In this study we explore a class of adjuvant that harnesses mucosal-associated invariant T (MAIT) cells. We show evidence that intranasal immunization of MAIT cell agonists co-administered with protein, including the spike receptor binding domain from SARS-CoV-2 virus and hemagglutinin from influenza virus, induce protective humoral immunity and immunoglobulin A production. MAIT cell adjuvant activity is mediated by CD40L-dependent activation of dendritic cells and subsequent priming of T follicular helper cells. In summary, we show that MAIT cells are promising vaccine targets that can be utilized as cellular adjuvants in mucosal vaccines.
Collapse
Affiliation(s)
- Theresa E Pankhurst
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Kaitlin H Buick
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Joshua L Lange
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Andrew J Marshall
- Ferrier Research Institute, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Kaileen R Button
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Olga R Palmer
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Kathryn J Farrand
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Isabelle Montgomerie
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Thomas W Bird
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Ngarangi C Mason
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Joanna Kuang
- Department of Microbiology and Immunology, University of Otago, Dunedin 9016, New Zealand
| | - Benjamin J Compton
- Ferrier Research Institute, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Davide Comoletti
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Mariolina Salio
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Vincenzo Cerundolo
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | | | - Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Lisa M Connor
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Malaghan Institute of Medical Research, Wellington 6242, New Zealand.
| |
Collapse
|
15
|
Pandey P, Kim SH, Subedi L, Mujahid K, Kim Y, Cho YC, Shim JH, Kim KT, Cho SS, Choi JU, Park JW. Oral lymphatic delivery of alpha-galactosylceramide and ovalbumin evokes anti-cancer immunization. J Control Release 2023; 356:507-524. [PMID: 36907564 DOI: 10.1016/j.jconrel.2023.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/16/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023]
Abstract
We developed an orally delivered nanoemulsion that induces cancer immunization. It consists of tumor antigen-loaded nano-vesicles carrying the potent invariant natural killer T-cell (iNKT) activator α-galactosylceramide (α-GalCer), to trigger cancer immunity by effectively activating both innate and adaptive immunity. It was validated that adding bile salts to the system boosted intestinal lymphatic transport as well as the oral bioavailability of ovalbumin (OVA) via the chylomicron pathway. To increase intestinal permeability further and amplify the antitumor responses, an ionic complex of cationic lipid 1,2-dioleyl-3-trimethylammonium propane (DTP) with sodium deoxycholate (DA) (DDP) and α-GalCer were anchored onto the outer oil layer to form OVA-NE#3. As expected, OVA-NE#3 exhibited tremendously improved intestinal cell permeability as well as enhanced delivery to mesenteric lymph nodes (MLNs). Subsequent activation of dendritic cells and iNKTs, in MLNs were also observed. Tumor growth in OVA-expressing mice with melanoma was more strongly suppressed (by 71%) after oral administration of OVA-NE#3 than in untreated controls, confirming the strong immune response induced by the system. The serum levels of OVA-specific IgG1 and IgG2a were 3.52- and 6.14-fold higher than in controls. Treating OVA-NE#3 increased the numbers of tumor-infiltrating lymphocytes, including cytotoxic T-cell and M1-like macrophage. Antigen- and α-GalCer-associated enrichment of dendritic cells and iNKTs in tumor tissues also increased after OVA-NE#3 treatment. These observations indicate that our system induces both cellular and humoral immunity by targeting the oral lymphatic system. It may offer a promising oral anti-cancer vaccination strategy that involves the induction of systemic anti-cancer immunization.
Collapse
Affiliation(s)
- Prashant Pandey
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Seung Hyun Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Laxman Subedi
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Khizra Mujahid
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yebon Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Young-Chang Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jung-Hyun Shim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Ki-Taek Kim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Seung-Sik Cho
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Jeong Uk Choi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Jin Woo Park
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea.
| |
Collapse
|
16
|
Umbreen H, Zhang X, Tang KT, Lin CC. Regulation of Myeloid Dendritic Cells by Synthetic and Natural Compounds for the Treatment of Rheumatoid Arthritis. Int J Mol Sci 2022; 24:ijms24010238. [PMID: 36613683 PMCID: PMC9820359 DOI: 10.3390/ijms24010238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Different subsets of dendritic cells (DCs) participate in the development of rheumatoid arthritis (RA). In particular, myeloid DCs play a key role in the generation of autoreactive T and B cells. Herein, we undertook a literature review on those synthetic and natural compounds that have therapeutic efficacy/potential for RA and act through the regulation of myeloid DCs. Most of these compounds inhibit both the maturation of DCs and their secretion of inflammatory cytokines and, subsequently, alter the downstream T-cell response (suppression of Th1 and Th17 responses while expanding the Treg response). The majority of the synthetic compounds are approved for the treatment of patients with RA, which is consistent with the importance of DCs in the pathogenesis of RA. All of the natural compounds are derived from plants. Their DC-modulating effect has been demonstrated both in vitro and in vivo. In addition, these natural products ameliorate arthritis in rodents and are potential therapeutics for human RA.
Collapse
Affiliation(s)
- Hira Umbreen
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Xiang Zhang
- Department of Molecular Medicine and Surgery, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Kuo-Tung Tang
- Division of Allergy, Immunology, and Rheumatology, Taichung Veterans General Hospital, Taichung 407, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Correspondence: (K.-T.T.); (C.-C.L.); Tel.: +886-4-23592525 (ext. 3334) (K.-T.T.); +886-4-23592525 (ext. 3003) (C.-C.L.); Fax: +886-4-23503285 (K.-T.T. & C.-C.L.)
| | - Chi-Chien Lin
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan
- Institute of Biomedical Science, The iEGG and Animal Biotechnology Center, National Chung-Hsing University, Taichung 402, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (K.-T.T.); (C.-C.L.); Tel.: +886-4-23592525 (ext. 3334) (K.-T.T.); +886-4-23592525 (ext. 3003) (C.-C.L.); Fax: +886-4-23503285 (K.-T.T. & C.-C.L.)
| |
Collapse
|
17
|
Role of NKT cells in cancer immunotherapy-from bench to bed. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:29. [PMID: 36460881 DOI: 10.1007/s12032-022-01888-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/08/2022] [Indexed: 12/04/2022]
Abstract
Natural killer T (NKT) cells are a specific T cell subset known to express the αβ-T cell receptor (TCR) for antigens identification and express typical NK cell specifications, such as surface expression of CD56 and CD16 markers as well as production of granzyme. Human NKT cells are divided into two subgroups based on their cytokine receptor and TCR repertoire. Both of them are CD1-restricted and recognize lipid antigens presented by CD1d molecules. Studies have demonstrated that these cells are essential in defense against malignancies. These cells secret proinflammatory and regulatory cytokines that stimulate or suppress immune system responses. In several murine tumor models, activation of type I NKT cells induces tumor rejection and inhibits metastasis's spread. However, type II NKT cells are associated with an inhibitory and regulatory function during tumor immune responses. Variant NKT cells may suppress tumor immunity via different mechanisms that require cross-talk with other immune-regulatory cells. NKT-like cells display high tumor-killing abilities against many tumor cells. In the recent decade, different studies have been performed based on the application of NKT-based immunotherapy for cancer therapy. Moreover, manipulation of NKT cells through administering autologous dendritic cell (DC) loaded with α-galactosylceramide (α-GalCer) and direct α-GalCer injection has also been tested. In this review, we described different subtypes of NKT cells, their function in the anti-tumor immune responses, and the application of NKT cells in cancer immunotherapy from bench to bed.
Collapse
|
18
|
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: 28] [Impact Index Per Article: 9.3] [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
|
19
|
Fujii SI, Kawamata T, Shimizu K, Nakabayashi J, Yamasaki S, Iyoda T, Shinga J, Nakazato H, Sanpei A, Kawamura M, Ueda S, Dörrie J, Mojsov S, Dhodapkar MV, Hidaka M, Nojima M, Nagamura F, Yoshida S, Goto T, Tojo A. Reinvigoration of innate and adaptive immunity via therapeutic cellular vaccine for patients with AML. Mol Ther Oncolytics 2022. [DOI: 10.1016/j.omto.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
20
|
Delfanti G, Cortesi F, Perini A, Antonini G, Azzimonti L, de Lalla C, Garavaglia C, Squadrito ML, Fedeli M, Consonni M, Sesana S, Re F, Shen H, Dellabona P, Casorati G. TCR-engineered iNKT cells induce robust antitumor response by dual targeting cancer and suppressive myeloid cells. Sci Immunol 2022; 7:eabn6563. [PMID: 35984893 DOI: 10.1126/sciimmunol.abn6563] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Adoptive immunotherapy with T cells engineered with tumor-specific T cell receptors (TCRs) holds promise for cancer treatment. However, suppressive cues generated in the tumor microenvironment (TME) can hinder the efficacy of these therapies, prompting the search for strategies to overcome these detrimental conditions and improve cellular therapeutic approaches. CD1d-restricted invariant natural killer T (iNKT) cells actively participate in tumor immunosurveillance by restricting suppressive myeloid populations in the TME. Here, we showed that harnessing iNKT cells with a second TCR specific for a tumor-associated peptide generated bispecific effectors for CD1d- and major histocompatibility complex (MHC)-restricted antigens in vitro. Upon in vivo transfer, TCR-engineered iNKT (TCR-iNKT) cells showed the highest efficacy in restraining the progression of multiple tumors that expressed the cognate antigen compared with nontransduced iNKT cells or CD8+ T cells engineered with the same TCR. TCR-iNKT cells achieved robust cancer control by simultaneously modulating intratumoral suppressive myeloid populations and killing malignant cells. This dual antitumor function was further enhanced when the iNKT cell agonist α-galactosyl ceramide (α-GalCer) was administered as a therapeutic booster through a platform that ensured controlled delivery at the tumor site, named multistage vector (MSV). These preclinical results support the combination of tumor-redirected TCR-iNKT cells and local α-GalCer boosting as a potential therapy for patients with cancer.
Collapse
Affiliation(s)
- Gloria Delfanti
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Filippo Cortesi
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Alessandra Perini
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Gaia Antonini
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan 20132, Italy
| | | | - Claudia de Lalla
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Claudio Garavaglia
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Mario L Squadrito
- Targeted Cancer Gene Therapy Unit, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan 20132, Italy
| | - Maya Fedeli
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Michela Consonni
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Silvia Sesana
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20854 Monza, Italy
| | - Francesca Re
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20854 Monza, Italy
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Paolo Dellabona
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Giulia Casorati
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan 20132, Italy
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Delfanti G, Dellabona P, Casorati G, Fedeli M. Adoptive Immunotherapy With Engineered iNKT Cells to Target Cancer Cells and the Suppressive Microenvironment. Front Med (Lausanne) 2022; 9:897750. [PMID: 35615083 PMCID: PMC9125179 DOI: 10.3389/fmed.2022.897750] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
Invariant Natural Killer T (iNKT) cells are T lymphocytes expressing a conserved semi-invariant TCR specific for lipid antigens (Ags) restricted for the monomorphic MHC class I-related molecule CD1d. iNKT cells infiltrate mouse and human tumors and play an important role in the immune surveillance against solid and hematological malignancies. Because of unique functional features, they are attractive platforms for adoptive cells immunotherapy of cancer compared to conventional T cells. iNKT cells can directly kill CD1d-expressing cancer cells, but also restrict immunosuppressive myelomonocytic populations in the tumor microenvironment (TME) via CD1d-cognate recognition, promoting anti-tumor responses irrespective of the CD1d expression by cancer cells. Moreover, iNKT cells can be adoptively transferred across MHC barriers without risk of alloreaction because CD1d molecules are identical in all individuals, in addition to their ability to suppress graft vs. host disease (GvHD) without impairing the anti-tumor responses. Within this functional framework, iNKT cells are successfully engineered to acquire a second antigen-specificity by expressing recombinant TCRs or Chimeric Antigen Receptor (CAR) specific for tumor-associated antigens, enabling the direct targeting of antigen-expressing cancer cells, while maintaining their CD1d-dependent functions. These new evidences support the exploitation of iNKT cells for donor unrestricted, and possibly off the shelf, adoptive cell therapies enabling the concurrent targeting of cancer cells and suppressive microenvironment.
Collapse
Affiliation(s)
- Gloria Delfanti
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- *Correspondence: Gloria Delfanti
| | - Paolo Dellabona
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Casorati
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Giulia Casorati
| | - Maya Fedeli
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Maya Fedeli
| |
Collapse
|
24
|
Hossian AKMN, Hackett CS, Brentjens RJ, Rafiq S. Multipurposing CARs: Same engine, different vehicles. Mol Ther 2022; 30:1381-1395. [PMID: 35151842 PMCID: PMC9077369 DOI: 10.1016/j.ymthe.2022.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/16/2021] [Accepted: 02/08/2022] [Indexed: 11/22/2022] Open
Abstract
T cells genetically engineered to recognize and eliminate tumor cells through synthetic chimeric antigen receptors (CARs) have demonstrated remarkable clinical efficacy against B cell leukemia over the past decade. This therapy is a form of highly personalized medicine that involves genetically modifying a patient's T cells to recognize and kill cancer cells. With the FDA approval of 5 CAR T cell products, this approach has been validated as a powerful new drug in the therapeutic armamentarium against cancer. Researchers are now studying how to expand this technology beyond its use in conventional polyclonal αβ T cells to address limitations to the current therapy in cancer and applications beyond it. Considering the specific characteristics of immune cell from diverse lineages, several preclinical and clinical studies are under way to assess the advantages of CAR-redirected function in these cells and apply the lessons learned from CAR T cell therapy in cancer to other diseases.
Collapse
Affiliation(s)
- A K M Nawshad Hossian
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
| | - Christopher S Hackett
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Renier J Brentjens
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA.
| | - Sarwish Rafiq
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
25
|
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
|
26
|
Abstract
Chronic inflammation increases the risk of several cancers, including gastric, colon, and hepatic cancers. Conversely, tumors, similar to tissue injury, trigger an inflammatory response coordinated by the innate immune system. Cellular and molecular mediators of inflammation modulate tumor growth directly and by influencing the adaptive immune response. Depending on the balance of immune cell types and signals within the tumor microenvironment, inflammation can support or restrain the tumor. Adding to the complexity, research from the past two decades has revealed that innate immune cells are highly heterogeneous and plastic, with variable phenotypes depending on tumor type, stage, and treatment. The field is now on the cusp of being able to harness this wealth of data to (a) classify tumors on the basis of their immune makeup, with implications for prognosis, treatment choice, and clinical outcome, and (b) design therapeutic strategies that activate antitumor immune responses by targeting innate immune cells.
Collapse
Affiliation(s)
- Laura Maiorino
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| | | | - Lijuan Sun
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA; , ,
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA; , ,
| |
Collapse
|
27
|
Lang GA, Norman K, Amadou Amani S, Shadid TM, Ballard JD, Lang ML. Use of a Clostridioides difficile Murine Immunization and Challenge Model to Evaluate Single and Combination Vaccine Adjuvants Consisting of Alum and NKT Cell-Activating Ligands. Front Immunol 2022; 12:818734. [PMID: 35095921 PMCID: PMC8794951 DOI: 10.3389/fimmu.2021.818734] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 12/27/2021] [Indexed: 01/14/2023] Open
Abstract
Adjuvant combinations may enhance or broaden the expression of immune responses to vaccine antigens. Information on whether established Alum type adjuvants can be combined with experimental CD1d ligand adjuvants is currently lacking. In this study, we used a murine Clostridioides difficile immunization and challenge model to evaluate Alum (Alhydrogel™), α-galactosylceramide (α-GC), and one of its analogs 7DW8-5 singly and in combination as vaccine adjuvants. We observed that the Alum/α-GC combination caused modest enhancement of vaccine antigen-specific IgG1 and IgG2b responses, and a broadening to include IgG2c that did not significantly impact overall protection. Similar observations were made using the Alum/7DW8-5 combination. Examination of the impact of adjuvants on NKT cells revealed expansion of invariant NKT (iNKT) cells with modest expansion of their iNKTfh subset and little effect on diverse NKT (dNKT) cells. Side effects of the adjuvants was determined and revealed transient hepatotoxicity when Alum/α-GC was used in combination but not singly. In summary these results showed that the Alum/α-GC or the Alum/7DW8-5 combination could exert distinct effects on the NKT cell compartment and on isotype switch to produce Th1-driven IgG subclasses in addition to Alum/Th2-driven subclasses. While Alum alone was efficacious in stimulating IgG-mediated protection, and α-GC offered no apparent additional benefit in the C. difficile challenge model, the work herein reveals immune response features that could be optimized and harnessed in other vaccine contexts.
Collapse
|
28
|
Akache B, Stark FC, Agbayani G, Renner TM, McCluskie MJ. Adjuvants: Engineering Protective Immune Responses in Human and Veterinary Vaccines. Methods Mol Biol 2022; 2412:179-231. [PMID: 34918246 DOI: 10.1007/978-1-0716-1892-9_9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Adjuvants are key components of many vaccines, used to enhance the level and breadth of the immune response to a target antigen, thereby enhancing protection from the associated disease. In recent years, advances in our understanding of the innate and adaptive immune systems have allowed for the development of a number of novel adjuvants with differing mechanisms of action. Herein, we review adjuvants currently approved for human and veterinary use, describing their use and proposed mechanisms of action. In addition, we will discuss additional promising adjuvants currently undergoing preclinical and/or clinical testing.
Collapse
Affiliation(s)
- Bassel Akache
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Felicity C Stark
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Gerard Agbayani
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Tyler M Renner
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Michael J McCluskie
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada.
| |
Collapse
|
29
|
Gao SY, Amaro-Mugridge NB, Madrid-Weiss J, Petkovic N, Vanegas N, Visvanathan K, Williams BRG, MacDiarmid JA, Brahmbhatt H. Nanocell COVID-19 vaccine triggers a novel immune response pathway producing high-affinity antibodies which neutralize all variants of concern. Front Immunol 2022; 13:1038562. [PMID: 36818474 PMCID: PMC9929940 DOI: 10.3389/fimmu.2022.1038562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/19/2022] [Indexed: 01/28/2023] Open
Abstract
Most current anti-viral vaccines elicit a humoral and cellular immune response via the pathway of phagocytic cell mediated viral antigen presentation to B and T cell surface receptors. However, this pathway results in reduced ability to neutralize S-protein Receptor Binding Domains (RBDs) from several Variants of Concern (VOC) and the rapid waning of memory B cell response requiring vaccine reformulation to cover dominant VOC S-proteins and multiple boosters. Here we show for the first time in mice and humans, that a bacterially derived, non-living, nanocell (EDV; EnGeneIC Dream Vector) packaged with plasmid expressed SARS-CoV-2 S-protein and α-galactosyl ceramide adjuvant (EDV-COVID-αGC), stimulates an alternate pathway due to dendritic cells (DC) displaying both S-polypeptides and αGC thereby recruiting and activating iNKT cells with release of IFNγ. This triggers DC activation/maturation, activation of follicular helper T cells (TFH), cognate help to B cells with secretion of a cytokine milieu promoting B cell maturation, somatic hypermutation in germinal centers to result in high affinity antibodies. Surrogate virus neutralization tests show 90-100% neutralization of ancestral and early VOC in mice and human trial volunteers. EDV-COVID-αGC as a third dose booster neutralized Omicron BA. 4/5. Serum and PBMC analyses reveal long lasting S-specific memory B and T cells. In contrast, control EDVs lacking αGC, did not engage the iNKT/DC pathway resulting in antibody responses unable to neutralize all VOCs and had a reduced B cell memory. The vaccine is lyophilized, stored and transported at room temperature with a shelf-life of over a year.
Collapse
Affiliation(s)
| | | | | | | | | | - Kumar Visvanathan
- Kumar Visvanathan, Department of Medicine, University of Melbourne, Fitzroy, VIC, Australia
| | - Bryan R G Williams
- Department of Molecular and Translational Science, Hudson Institute of Medical Research, Monash University Faculty of Medicine, Nursing and Health Sciences, Clayton, VIC, Australia
| | | | | |
Collapse
|
30
|
Fujii SI, Yamasaki S, Hanada K, Ueda S, Kawamura M, Shimizu K. Cancer immunotherapy using artificial adjuvant vector cells to deliver NY-ESO-1 antigen to dendritic cells in situ. Cancer Sci 2021; 113:864-874. [PMID: 34971473 PMCID: PMC8898705 DOI: 10.1111/cas.15259] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/19/2021] [Accepted: 12/22/2021] [Indexed: 11/26/2022] Open
Abstract
NY‐ESO‐1 is a cancer/testis antigen expressed in various cancer types. However, the induction of NY‐ESO‐1‐specific CTLs through vaccines is somewhat difficult. Thus, we developed a new type of artificial adjuvant vector cell (aAVC‐NY‐ESO‐1) expressing a CD1d‐NKT cell ligand complex and a tumor‐associated antigen, NY‐ESO‐1. First, we determined the activation of invariant natural killer T (iNKT) and natural killer (NK) cell responses by aAVC‐NY‐ESO‐1. We then showed that the NY‐ESO‐1‐specific CTL response was successfully elicited through aAVC‐NY‐ESO‐1 therapy. After injection of aAVC‐NY‐ESO‐1, we found that dendritic cells (DCs) in situ expressed high levels of costimulatory molecules and produced interleukn‐12 (IL‐12), indicating that DCs undergo maturation in vivo. Furthermore, the NY‐ESO‐1 antigen from aAVC‐NY‐ESO‐1 was delivered to the DCs in vivo, and it was presented on MHC class I molecules. The cross‐presentation of the NY‐ESO‐1 antigen was absent in conventional DC‐deficient mice, suggesting a host DC‐mediated CTL response. Thus, this strategy helps generate sufficient CD8+ NY‐ESO‐1‐specific CTLs along with iNKT and NK cell activation, resulting in a strong antitumor effect. Furthermore, we established a human DC‐transferred NOD/Shi‐scid/IL‐2γcnull immunodeficient mouse model and showed that the NY‐ESO‐1 antigen from aAVC‐NY‐ESO‐1 was cross‐presented to antigen‐specific CTLs through human DCs. Taken together, these data suggest that aAVC‐NY‐ESO‐1 has potential for harnessing innate and adaptive immunity against NY‐ESO‐1‐expressing malignancies.
Collapse
Affiliation(s)
- Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Research Center for Integrative Medicine (IMS), Yokohama, Japan.,RIKEN Program for drug discovery and medical technology platforms, Yokohama, Japan
| | - Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Research Center for Integrative Medicine (IMS), Yokohama, Japan
| | - Kenichi Hanada
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shogo Ueda
- Laboratory for Immunotherapy, RIKEN Research Center for Integrative Medicine (IMS), Yokohama, Japan
| | - Masami Kawamura
- Laboratory for Immunotherapy, RIKEN Research Center for Integrative Medicine (IMS), Yokohama, Japan
| | - Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Research Center for Integrative Medicine (IMS), Yokohama, Japan
| |
Collapse
|
31
|
Zimmerer JM, Ringwald BA, Chaudhari SR, Han J, Peterson CM, Warren RT, Hart MM, Abdel-Rasoul M, Bumgardner GL. Invariant NKT Cells Promote the Development of Highly Cytotoxic Multipotent CXCR3 +CCR4 +CD8 + T Cells That Mediate Rapid Hepatocyte Allograft Rejection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:3107-3121. [PMID: 34810223 PMCID: PMC9124232 DOI: 10.4049/jimmunol.2100334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 10/13/2021] [Indexed: 12/22/2022]
Abstract
Hepatocyte transplant represents a treatment for metabolic disorders but is limited by immunogenicity. Our prior work identified the critical role of CD8+ T cells, with or without CD4+ T cell help, in mediating hepatocyte rejection. In this study, we evaluated the influence of invariant NKT (iNKT) cells, uniquely abundant in the liver, upon CD8-mediated immune responses in the presence and absence of CD4+ T cells. To investigate this, C57BL/6 (wild-type) and iNKT-deficient Jα18 knockout mice (cohorts CD4 depleted) were transplanted with allogeneic hepatocytes. Recipients were evaluated for alloprimed CD8+ T cell subset composition, allocytotoxicity, and hepatocyte rejection. We found that CD8-mediated allocytotoxicity was significantly decreased in iNKT-deficient recipients and was restored by adoptive transfer of iNKT cells. In the absence of both iNKT cells and CD4+ T cells, CD8-mediated allocytotoxicity and hepatocyte rejection was abrogated. iNKT cells enhance the proportion of a novel subset of multipotent, alloprimed CXCR3+CCR4+CD8+ cytolytic T cells that develop after hepatocyte transplant and are abundant in the liver. Alloprimed CXCR3+CCR4+CD8+ T cells express cytotoxic effector molecules (perforin/granzyme and Fas ligand) and are distinguished from alloprimed CXCR3+CCR4-CD8+ T cells by a higher proportion of cells expressing TNF-α and IFN-γ. Furthermore, alloprimed CXCR3+CCR4+CD8+ T cells mediate higher allocytotoxicity and more rapid allograft rejection. Our data demonstrate the important role of iNKT cells in promoting the development of highly cytotoxic, multipotent CXCR3+CCR4+CD8+ T cells that mediate rapid rejection of allogeneic hepatocytes engrafted in the liver. Targeting iNKT cells may be an efficacious therapy to prevent rejection of intrahepatic cellular transplants.
Collapse
Affiliation(s)
- Jason M Zimmerer
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Bryce A Ringwald
- Medical Student Research Program, The Ohio State University College of Medicine, Columbus, OH
| | - Sachi R Chaudhari
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Jing Han
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH; and
| | - Chelsea M Peterson
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Robert T Warren
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Madison M Hart
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | | | - Ginny L Bumgardner
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH;
| |
Collapse
|
32
|
Cianga VA, Dănăilă CD, Antohe I, Oană R, Mențel M, Ivanov I, Dragoș L, Dăscălescu AS. A very rare case of FLT3-D835 positive blastic plasmacytoid dendritic cell neoplasm. Arch Clin Cases 2021; 7:57-62. [PMID: 34754929 PMCID: PMC8565708 DOI: 10.22551/2020.29.0704.10174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Blastic plasmacytoid dendritic cell neoplasms (BPDCNs) are extremely rare and aggressive hematological malignancies that derive from precursors of plasmacytoid dendritic cells (pDC) and frequently involve skin lesions and bone marrow infiltration. They mostly affect the elderly population and the prognosis is poor with the therapeutic choices currently available. Diagnosis is made with the help of tools such as immunohistochemistry and flow cytometry. Here, we present a particular case of BPDCN with a positive FLT3-D835 mutation and we discuss the possible impact this may have on the evolution of the disease and response to treatment.
Collapse
Affiliation(s)
- Vlad Andrei Cianga
- Grigore T. Popa University of Medicine and Pharmacy, Iași, Romania.,Department of Clinical Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Cătălin Doru Dănăilă
- Grigore T. Popa University of Medicine and Pharmacy, Iași, Romania.,Department of Clinical Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Ion Antohe
- Department of Clinical Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Raluca Oană
- Department of Cytology, Regional Institute of Oncology, Iasi, Romania
| | - Mihaela Mențel
- Department of Immunophenotyping, Regional Institute of Oncology, Iasi, Romania
| | - Iuliu Ivanov
- Department of Molecular Diagnostics, Regional Institute of Oncology, Iasi, Romania
| | - Loredana Dragoș
- Department of Molecular Diagnostics, Regional Institute of Oncology, Iasi, Romania
| | - Angela Smaranda Dăscălescu
- Grigore T. Popa University of Medicine and Pharmacy, Iași, Romania.,Department of Clinical Hematology, Regional Institute of Oncology, Iasi, Romania
| |
Collapse
|
33
|
Simonetta F, Lohmeyer JK, Hirai T, Maas-Bauer K, Alvarez M, Wenokur AS, Baker J, Aalipour A, Ji X, Haile S, Mackall CL, Negrin RS. Allogeneic CAR Invariant Natural Killer T Cells Exert Potent Antitumor Effects through Host CD8 T-Cell Cross-Priming. Clin Cancer Res 2021; 27:6054-6064. [PMID: 34376537 PMCID: PMC8563377 DOI: 10.1158/1078-0432.ccr-21-1329] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/13/2021] [Accepted: 07/30/2021] [Indexed: 01/09/2023]
Abstract
PURPOSE The development of allogeneic chimeric antigen receptor (CAR) T-cell therapies for off-the-shelf use is a major goal that faces two main immunologic challenges, namely the risk of graft-versus-host disease (GvHD) induction by the transferred cells and the rejection by the host immune system limiting their persistence. In this work we assessed the direct and indirect antitumor effect of allogeneic CAR-engineered invariant natural killer T (iNKT) cells, a cell population without GvHD-induction potential that displays immunomodulatory properties. EXPERIMENTAL DESIGN After assessing murine CAR iNKT cells direct antitumor effects in vitro and in vivo, we employed an immunocompetent mouse model of B-cell lymphoma to assess the interaction between allogeneic CAR iNKT cells and endogenous immune cells. RESULTS We demonstrate that allogeneic CAR iNKT cells exerted potent direct and indirect antitumor activity when administered across major MHC barriers by inducing tumor-specific antitumor immunity through host CD8 T-cell cross-priming. CONCLUSIONS In addition to their known direct cytotoxic effect, allogeneic CAR iNKT cells induce host CD8 T-cell antitumor responses, resulting in a potent antitumor effect lasting longer than the physical persistence of the allogeneic cells. The utilization of off-the-shelf allogeneic CAR iNKT cells could meet significant unmet needs in the clinic.
Collapse
Affiliation(s)
- Federico Simonetta
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
- Division of Hematology, Department of Oncology, Geneva University Hospitals and Translational Research Centre in Onco-Haematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Juliane K Lohmeyer
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Toshihito Hirai
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Kristina Maas-Bauer
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Maite Alvarez
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Arielle S Wenokur
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Jeanette Baker
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Amin Aalipour
- Department of Bioengineering, Stanford University School of Medicine, Stanford, California
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Xuhuai Ji
- Human Immune Monitoring Center, Stanford University School of Medicine, Stanford, California
| | - Samuel Haile
- Department of Pediatrics, Stanford University, Stanford, California
| | - Crystal L Mackall
- Department of Pediatrics, Stanford University, Stanford, California
- Stanford Cancer Institute, Stanford University, Stanford, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Robert S Negrin
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
34
|
Nelson A, Lukacs JD, Johnston B. The Current Landscape of NKT Cell Immunotherapy and the Hills Ahead. Cancers (Basel) 2021; 13:cancers13205174. [PMID: 34680322 PMCID: PMC8533824 DOI: 10.3390/cancers13205174] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Natural killer T (NKT) cells are a subset of lipid-reactive T cells that enhance anti-tumor immunity. While preclinical studies have shown NKT cell immunotherapy to be safe and effective, clinical studies lack predictable therapeutic efficacy and no approved treatments exist. In this review, we outline the current strategies, challenges, and outlook for NKT cell immunotherapy. Abstract NKT cells are a specialized subset of lipid-reactive T lymphocytes that play direct and indirect roles in immunosurveillance and anti-tumor immunity. Preclinical studies have shown that NKT cell activation via delivery of exogenous glycolipids elicits a significant anti-tumor immune response. Furthermore, infiltration of NKT cells is associated with a good prognosis in several cancers. In this review, we aim to summarize the role of NKT cells in cancer as well as the current strategies and status of NKT cell immunotherapy. This review also examines challenges and future directions for improving the therapy.
Collapse
Affiliation(s)
- Adam Nelson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (A.N.); (J.D.L.)
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 4R2, Canada
| | - Jordan D. Lukacs
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (A.N.); (J.D.L.)
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 4R2, Canada
| | - Brent Johnston
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (A.N.); (J.D.L.)
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 4R2, Canada
- Department of Pediatrics, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Correspondence:
| |
Collapse
|
35
|
α-Galactosylceramide-Reactive NKT Cells Increase IgG1 Class Switch against a Clostridioides difficile Polysaccharide Antigen and Enhance Immunity against a Live Pathogen Challenge. Infect Immun 2021; 89:e0043821. [PMID: 34424751 DOI: 10.1128/iai.00438-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
All clinical Clostridioides difficile strains identified to date express a surface capsule-like polysaccharide structure known as polysaccharide II (PSII). The PSII antigen is immunogenic and, when conjugated to a protein carrier, induces a protective antibody response in animal models. Given that CD1d-restricted natural killer T (NKT) cells promote antibody responses, including those against carbohydrates, we tested the hypothesis that immunization with PSII and a CD1d-binding glycolipid adjuvant could lead to enhanced protection against a live C. difficile challenge. We purified PSII from a clinical isolate of C. difficile and immunized B6 mice with PSII alone or PSII plus the CD1d-binding glycolipid α-galactosylceramide (α-GC). PSII-specific IgM and IgG titers were evident in sera from immunized mice. The inclusion of α-GC had a modest influence on isotype switch but increased the IgG1/IgG2c ratio. Enhanced protection against C. difficile disease was achieved by inclusion of the α-GC ligand and was associated with reduced bacterial numbers in fecal pellets. In contrast, NKT-deficient Traj18-/- mice were not protected by the PSII/α-GC immunization modality. Absence of NKT cells similarly had a modest effect on isotype switch, but ratios of IgG1/IgG2c decreased. These results indicate that α-GC-driven NKT cells move the humoral immune response against C. difficile PSII antigen toward Th2-driven IgG1 and may contribute to augmented protection. This study suggests that NKT activation represents a pathway for additional B-cell help that could be used to supplement existing efforts to develop vaccines against polysaccharides derived from C. difficile and other pathogens.
Collapse
|
36
|
Brettschneider EES, Terabe M. The Role of NKT Cells in Glioblastoma. Cells 2021; 10:cells10071641. [PMID: 34208864 PMCID: PMC8307781 DOI: 10.3390/cells10071641] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma is an aggressive and deadly cancer, but to date, immunotherapies have failed to make significant strides in improving prognoses for glioblastoma patients. One of the current challenges to developing immunological interventions for glioblastoma is our incomplete understanding of the numerous immunoregulatory mechanisms at play in the glioblastoma tumor microenvironment. We propose that Natural Killer T (NKT) cells, which are unconventional T lymphocytes that recognize lipid antigens presented by CD1d molecules, may play a key immunoregulatory role in glioblastoma. For example, evidence suggests that the activation of type I NKT cells can facilitate anti-glioblastoma immune responses. On the other hand, type II NKT cells are known to play an immunosuppressive role in other cancers, as well as to cross-regulate type I NKT cell activity, although their specific role in glioblastoma remains largely unclear. This review provides a summary of our current understanding of NKT cells in the immunoregulation of glioblastoma as well as highlights the involvement of NKT cells in other cancers and central nervous system diseases.
Collapse
Affiliation(s)
- Emily E. S. Brettschneider
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA;
- Nuffield Department of Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford OX3 7DQ, UK
| | - Masaki Terabe
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA;
- Correspondence: ; Tel.: +1-240-760-6731
| |
Collapse
|
37
|
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
|
38
|
Kyaw T, Loveland P, Kanellakis P, Cao A, Kallies A, Huang AL, Peter K, Toh BH, Bobik A. Alarmin-activated B cells accelerate murine atherosclerosis after myocardial infarction via plasma cell-immunoglobulin-dependent mechanisms. Eur Heart J 2021; 42:938-947. [PMID: 33338208 DOI: 10.1093/eurheartj/ehaa995] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/30/2020] [Accepted: 11/29/2020] [Indexed: 12/25/2022] Open
Abstract
AIMS Myocardial infarction (MI) accelerates atherosclerosis and greatly increases the risk of recurrent cardiovascular events for many years, in particular, strokes and MIs. Because B cell-derived autoantibodies produced in response to MI also persist for years, we investigated the role of B cells in adaptive immune responses to MI. METHODS AND RESULTS We used an apolipoprotein-E-deficient (ApoE-/-) mouse model of MI-accelerated atherosclerosis to assess the importance of B cells. One week after inducing MI in atherosclerotic mice, we depleted B cells using an anti-CD20 antibody. This treatment prevented subsequent immunoglobulin G accumulation in plaques and MI-induced accelerated atherosclerosis. In gain of function experiments, we purified spleen B cells from mice 1 week after inducing MI and transferred these cells into atherosclerotic ApoE-/- mice, which greatly increased immunoglobulin G (IgG) accumulation in plaque and accelerated atherosclerosis. These B cells expressed many cytokines that promote humoural immunity and in addition, they formed germinal centres within the spleen where they differentiated into antibody-producing plasma cells. Specifically deleting Blimp-1 in B cells, the transcriptional regulator that drives their terminal differentiation into antibody-producing plasma cells prevented MI-accelerated atherosclerosis. Alarmins released from infarcted hearts were responsible for activating B cells via toll-like receptors and deleting MyD88, the canonical adaptor protein for inflammatory signalling downstream of toll-like receptors, prevented B-cell activation and MI-accelerated atherosclerosis. CONCLUSION Our data implicate early B-cell activation and autoantibodies as a central cause for accelerated atherosclerosis post-MI and identifies novel therapeutic strategies towards preventing recurrent cardiovascular events such as MI and stroke.
Collapse
Affiliation(s)
- Tin Kyaw
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Paula Loveland
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Peter Kanellakis
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Anh Cao
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Axel Kallies
- Department of Microbiology and Immunology, University of Melbourne, 792 Elizabeth Street, Melbourne, Vic 3000, Australia
| | - Alex L Huang
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Department of Cardiology, Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Department of Cardiology, Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia.,Department of Immunology, Central Clinical School, 99 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Ban-Hock Toh
- Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Alex Bobik
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia.,Department of Immunology, Central Clinical School, 99 Commercial Rd, Melbourne, VIC 3004, Australia
| |
Collapse
|
39
|
Ingram Z, Madan S, Merchant J, Carter Z, Gordon Z, Carey G, Webb TJ. Targeting Natural Killer T Cells in Solid Malignancies. Cells 2021; 10:1329. [PMID: 34072042 PMCID: PMC8227159 DOI: 10.3390/cells10061329] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/19/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022] Open
Abstract
Natural killer T (NKT) cells are a unique subset of lymphocytes that recognize lipid antigens in the context of the non-classical class I MHC molecule, CD1d, and serve as a link between the innate and adaptive immune system through their expeditious release of cytokines. Whereas NKT have well-established roles in mitigating a number of human diseases, herein, we focus on their role in cancer. NKT cells have been shown to directly and indirectly mediate anti-tumor immunity and manipulating their effector functions can have therapeutic significances in treatment of cancer. In this review, we highlight several therapeutic strategies that have been used to harness the effector functions of NKT cells to target different types of solid tumors. We also discuss several barriers to the successful utilization of NKT cells and summarize effective strategies being developed to harness the unique strengths of this potent population of T cells. Collectively, studies investigating the therapeutic potential of NKT cells serve not only to advance our understanding of this powerful immune cell subset, but also pave the way for future treatments focused on the modulation of NKT cell responses to enhance cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tonya J. Webb
- Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (Z.I.); (S.M.); (J.M.); (Z.C.); (Z.G.); (G.C.)
| |
Collapse
|
40
|
Nagy NA, de Haas AM, Geijtenbeek TBH, van Ree R, Tas SW, van Kooyk Y, de Jong EC. Therapeutic Liposomal Vaccines for Dendritic Cell Activation or Tolerance. Front Immunol 2021; 12:674048. [PMID: 34054859 PMCID: PMC8155586 DOI: 10.3389/fimmu.2021.674048] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DCs) are paramount in initiating and guiding immunity towards a state of activation or tolerance. This bidirectional capacity of DCs sets them at the center stage for treatment of cancer and autoimmune or allergic conditions. Accordingly, many clinical studies use ex vivo DC vaccination as a strategy to boost anti-tumor immunity or to suppress immunity by including vitamin D3, NF-κB inhibitors or retinoic acid to create tolerogenic DCs. As harvesting DCs from patients and differentiating these cells in vitro is a costly and cumbersome process, in vivo targeting of DCs has huge potential as nanoparticulate platforms equipped with activating or tolerogenic adjuvants can modulate DCs in their natural environment. There is a rapid expansion of the choices of nanoparticles and activation- or tolerance-promoting adjuvants for a therapeutic vaccine platform. In this review we highlight the most recent nanomedical approaches aimed at inducing immune activation or tolerance via targeting DCs, together with novel fundamental insights into the mechanisms inherent to fostering anti-tumor or tolerogenic immunity.
Collapse
Affiliation(s)
- Noémi Anna Nagy
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Aram M. de Haas
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Teunis B. H. Geijtenbeek
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald van Ree
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
- Department of Otorhinolaryngology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W. Tas
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Amsterdam Rheumatology and Immunology Center, University of Amsterdam, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Esther C. de Jong
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
41
|
Dölen Y, Gileadi U, Chen JL, Valente M, Creemers JHA, Van Dinther EAW, van Riessen NK, Jäger E, Hruby M, Cerundolo V, Diken M, Figdor CG, de Vries IJM. PLGA Nanoparticles Co-encapsulating NY-ESO-1 Peptides and IMM60 Induce Robust CD8 and CD4 T Cell and B Cell Responses. Front Immunol 2021; 12:641703. [PMID: 33717196 PMCID: PMC7947615 DOI: 10.3389/fimmu.2021.641703] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/28/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor-specific neoantigens can be highly immunogenic, but their identification for each patient and the production of personalized cancer vaccines can be time-consuming and prohibitively expensive. In contrast, tumor-associated antigens are widely expressed and suitable as an off the shelf immunotherapy. Here, we developed a PLGA-based nanoparticle vaccine that contains both the immunogenic cancer germline antigen NY-ESO-1 and an α-GalCer analog IMM60, as a novel iNKT cell agonist and dendritic cell transactivator. Three peptide sequences (85-111, 117-143, and 157-165) derived from immunodominant regions of NY-ESO-1 were selected. These peptides have a wide HLA coverage and were efficiently processed and presented by dendritic cells via various HLA subtypes. Co-delivery of IMM60 enhanced CD4 and CD8 T cell responses and antibody levels against NY-ESO-1 in vivo. Moreover, the nanoparticles have negligible systemic toxicity in high doses, and they could be produced according to GMP guidelines. Together, we demonstrated the feasibility of producing a PLGA-based nanovaccine containing immunogenic peptides and an iNKT cell agonist, that is activating DCs to induce antigen-specific T cell responses.
Collapse
Affiliation(s)
- Yusuf Dölen
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands.,Oncode Institute, Nijmegen, Netherlands
| | - Uzi Gileadi
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Ji-Li Chen
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Michael Valente
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands.,Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Jeroen H A Creemers
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands.,Oncode Institute, Nijmegen, Netherlands
| | - Eric A W Van Dinther
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands.,Oncode Institute, Nijmegen, Netherlands
| | - N Koen van Riessen
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Eliezer Jäger
- Institute of Macromolecular Chemistry v. v. i., Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Martin Hruby
- Institute of Macromolecular Chemistry v. v. i., Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Vincenzo Cerundolo
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Mustafa Diken
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands.,Oncode Institute, Nijmegen, Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| |
Collapse
|
42
|
Ghilas S, Enders MH, May R, Holz LE, Fernandez-Ruiz D, Cozijnsen A, Mollard V, Cockburn IA, McFadden GI, Heath WR, Beattie L. Development of Plasmodium-specific liver-resident memory CD8 + T cells after heat-killed sporozoite immunization in mice. Eur J Immunol 2021; 51:1153-1165. [PMID: 33486759 DOI: 10.1002/eji.202048757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 11/23/2020] [Accepted: 01/21/2021] [Indexed: 11/09/2022]
Abstract
Malaria remains a major cause of mortality in the world and an efficient vaccine is the best chance of reducing the disease burden. Vaccination strategies for the liver stage of disease that utilise injection of live radiation-attenuated sporozoites (RAS) confer sterile immunity, which is mediated by CD8+ memory T cells, with liver-resident memory T cells (TRM ) being particularly important. We have previously described a TCR transgenic mouse, termed PbT-I, where all CD8+ T cells recognize a specific peptide from Plasmodium. PbT-I form liver TRM cells upon RAS injection and are capable of protecting mice against challenge infection. Here, we utilize this transgenic system to examine whether nonliving sporozoites, killed by heat treatment (HKS), could trigger the development of Plasmodium-specific liver TRM cells. We found that HKS vaccination induced the formation of memory CD8+ T cells in the spleen and liver, and importantly, liver TRM cells were fewer in number than that induced by RAS. Crucially, we showed the number of TRM cells was significantly higher when HKS were combined with the glycolipid α-galactosylceramide as an adjuvant. In the future, this work could lead to development of an antimalaria vaccination strategy that does not require live sporozoites, providing greater utility.
Collapse
Affiliation(s)
- Sonia Ghilas
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Matthias H Enders
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Rose May
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Lauren E Holz
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Daniel Fernandez-Ruiz
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Anton Cozijnsen
- School of BioSciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Vanessa Mollard
- School of BioSciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ian A Cockburn
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2600, Australia
| | - Geoffrey I McFadden
- School of BioSciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - William R Heath
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Lynette Beattie
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
43
|
Wang L, Wu Q, Liu J, Zhang H, Bai L. Lactic acid inhibits iNKT cell functions via a phosphodiesterase-5 dependent pathway. Biochem Biophys Res Commun 2021; 547:9-14. [PMID: 33588236 DOI: 10.1016/j.bbrc.2021.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/03/2021] [Indexed: 11/15/2022]
Abstract
Lactic acid in tumor microenvironment inhibits iNKT cell functions and thus dampens their anti-tumor efficacy. The underlying mechanisms remain unclear. Here, we show that phosphodiesterase-5 inhibitors, sildenafil and tadalafil, promote IFN-γ and IL-4 production in iNKT cells in a cGMP-PKG pathway dependent manner. To favor their cytokine production, iNKT cells reduce Pde5a mRNA lever after activation. In line with the reduction of cytokines caused by lactic acid, lactic acid elevates Pde5a mRNA lever in activated iNKT cells. As a result, phosphodiesterase-5 inhibitor partially restores the cytokine production in lactic acid-treated cells. Our results demonstrate that phosphodiesterase-5 inhibits cytokine production in iNKT cells, and that contributes to the lactic acid-caused dysfunction of iNKT cells.
Collapse
Affiliation(s)
- Lili Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China
| | - Qielan Wu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China
| | - Jiwei Liu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China
| | - Huimin Zhang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China.
| | - Li Bai
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China.
| |
Collapse
|
44
|
Holz LE, Chua YC, de Menezes MN, Anderson RJ, Draper SL, Compton BJ, Chan STS, Mathew J, Li J, Kedzierski L, Wang Z, Beattie L, Enders MH, Ghilas S, May R, Steiner TM, Lange J, Fernandez-Ruiz D, Valencia-Hernandez AM, Osmond TL, Farrand KJ, Seneviratna R, Almeida CF, Tullett KM, Bertolino P, Bowen DG, Cozijnsen A, Mollard V, McFadden GI, Caminschi I, Lahoud MH, Kedzierska K, Turner SJ, Godfrey DI, Hermans IF, Painter GF, Heath WR. Glycolipid-peptide vaccination induces liver-resident memory CD8 + T cells that protect against rodent malaria. Sci Immunol 2021; 5:5/48/eaaz8035. [PMID: 32591409 DOI: 10.1126/sciimmunol.aaz8035] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 05/22/2020] [Indexed: 12/29/2022]
Abstract
Liver resident-memory CD8+ T cells (TRM cells) can kill liver-stage Plasmodium-infected cells and prevent malaria, but simple vaccines for generating this important immune population are lacking. Here, we report the development of a fully synthetic self-adjuvanting glycolipid-peptide conjugate vaccine designed to efficiently induce liver TRM cells. Upon cleavage in vivo, the glycolipid-peptide conjugate vaccine releases an MHC I-restricted peptide epitope (to stimulate Plasmodium-specific CD8+ T cells) and an adjuvant component, the NKT cell agonist α-galactosylceramide (α-GalCer). A single dose of this vaccine in mice induced substantial numbers of intrahepatic malaria-specific CD8+ T cells expressing canonical markers of liver TRM cells (CD69, CXCR6, and CD101), and these cells could be further increased in number upon vaccine boosting. We show that modifications to the peptide, such as addition of proteasomal-cleavage sequences or epitope-flanking sequences, or the use of alternative conjugation methods to link the peptide to the glycolipid improved liver TRM cell generation and led to the development of a vaccine able to induce sterile protection in C57BL/6 mice against Plasmodium berghei sporozoite challenge after a single dose. Furthermore, this vaccine induced endogenous liver TRM cells that were long-lived (half-life of ~425 days) and were able to maintain >90% sterile protection to day 200. Our findings describe an ideal synthetic vaccine platform for generating large numbers of liver TRM cells for effective control of liver-stage malaria and, potentially, a variety of other hepatotropic infections.
Collapse
Affiliation(s)
- Lauren E Holz
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Yu Cheng Chua
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Maria N de Menezes
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Regan J Anderson
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Sarah L Draper
- 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
| | - Susanna T S Chan
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Juby Mathew
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Jasmine Li
- Department of Microbiology, Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Zhongfang Wang
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Lynette Beattie
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Matthias H Enders
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia.,LIMES Institute, University of Bonn, Bonn, Germany
| | - Sonia Ghilas
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Rose May
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Thiago M Steiner
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Joshua Lange
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Daniel Fernandez-Ruiz
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Ana Maria Valencia-Hernandez
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Taryn L Osmond
- Malaghan Institute of Medical Research, Wellington, New Zealand.,Avalia Immunotherapies Limited, Lower Hutt, New Zealand
| | | | - Rebecca Seneviratna
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Catarina F Almeida
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Kirsteen M Tullett
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Patrick Bertolino
- Centenary Institute, The University of Sydney and AW Morrow Gastroenterology and Liver Centre, Liver Immunology Program, Newtown, NSW, Australia
| | - David G Bowen
- Centenary Institute, The University of Sydney and AW Morrow Gastroenterology and Liver Centre, Liver Immunology Program, Newtown, NSW, Australia
| | - Anton Cozijnsen
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | - Vanessa Mollard
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | | | - Irina Caminschi
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Mireille H Lahoud
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Turner
- Department of Microbiology, Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand. .,Avalia Immunotherapies Limited, Lower Hutt, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Wellington, New Zealand
| | - Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand. .,Avalia Immunotherapies Limited, Lower Hutt, New Zealand
| | - William R Heath
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia. .,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
45
|
Grabowska J, Stolk DA, Nijen Twilhaar MK, Ambrosini M, Storm G, van der Vliet HJ, de Gruijl TD, van Kooyk Y, den Haan JM. Liposomal Nanovaccine Containing α-Galactosylceramide and Ganglioside GM3 Stimulates Robust CD8 + T Cell Responses via CD169 + Macrophages and cDC1. Vaccines (Basel) 2021; 9:vaccines9010056. [PMID: 33467048 PMCID: PMC7830461 DOI: 10.3390/vaccines9010056] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/05/2021] [Accepted: 01/10/2021] [Indexed: 02/06/2023] Open
Abstract
Successful anti-cancer vaccines aim to prime and reinvigorate cytotoxic T cells and should therefore comprise a potent antigen and adjuvant. Antigen targeting to splenic CD169+ macrophages was shown to induce robust CD8+ T cell responses via antigen transfer to cDC1. Interestingly, CD169+ macrophages can also activate type I natural killer T-cells (NKT). NKT activation via ligands such as α-galactosylceramide (αGC) serve as natural adjuvants through dendritic cell activation. Here, we incorporated ganglioside GM3 and αGC in ovalbumin (OVA) protein-containing liposomes to achieve both CD169+ targeting and superior DC activation. The systemic delivery of GM3-αGC-OVA liposomes resulted in specific uptake by splenic CD169+ macrophages, stimulated strong IFNγ production by NKT and NK cells and coincided with the maturation of cDC1 and significant IL-12 production. Strikingly, superior induction of OVA-specific CD8+ T cells was detected after immunization with GM3-αGC-OVA liposomes. CD8+ T cell activation, but not B cell activation, was dependent on CD169+ macrophages and cDC1, while activation of NKT and NK cells were partially mediated by cDC1. In summary, GM3-αGC antigen-containing liposomes are a potent vaccination platform that promotes the interaction between different immune cell populations, resulting in strong adaptive immunity and therefore emerge as a promising anti-cancer vaccination strategy.
Collapse
Affiliation(s)
- Joanna Grabowska
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (J.G.); (D.A.S.); (M.K.N.T.); (M.A.); (Y.v.K.)
| | - Dorian A. Stolk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (J.G.); (D.A.S.); (M.K.N.T.); (M.A.); (Y.v.K.)
| | - Maarten K. Nijen Twilhaar
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (J.G.); (D.A.S.); (M.K.N.T.); (M.A.); (Y.v.K.)
| | - Martino Ambrosini
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (J.G.); (D.A.S.); (M.K.N.T.); (M.A.); (Y.v.K.)
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
- Department of Biomaterials Science and Technology, University of Twente, 7500 AE Enschede, The Netherlands
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Hans J. van der Vliet
- Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (H.J.v.d.V.); (T.D.d.G.)
- Lava Therapeutics, 3584 CM Utrecht, The Netherlands
| | - Tanja D. de Gruijl
- Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (H.J.v.d.V.); (T.D.d.G.)
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (J.G.); (D.A.S.); (M.K.N.T.); (M.A.); (Y.v.K.)
| | - Joke M.M. den Haan
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (J.G.); (D.A.S.); (M.K.N.T.); (M.A.); (Y.v.K.)
- Correspondence: ; Tel.: +31-20-4448080
| |
Collapse
|
46
|
Painter GF, Burn OK, Hermans IF. Using agonists for iNKT cells in cancer therapy. Mol Immunol 2020; 130:1-6. [PMID: 33340930 DOI: 10.1016/j.molimm.2020.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/02/2020] [Indexed: 01/03/2023]
Abstract
The capacity of α-galactosylceramide (α-GalCer) to act as an anti-cancer agent in mice through the specific stimulation of type I NKT (iNKT) cells has prompted extensive investigation to translate this finding to the clinic. However, low frequencies of iNKT cells in cancer patients and their hypo-responsiveness to repeated stimulation have been seen as barriers to its efficacy. Currently the most promising clinical application of α-GalCer, or its derivatives, is as stimuli for ex vivo expansion of iNKT cells for adoptive therapy, although some encouraging clinical results have recently been reported using α-GalCer pulsed onto large numbers of antigen presenting cells (APCs). In on-going preclinical studies, attempts to improve efficacy of injected iNKT cell agonists have focussed on optimising presentation in vivo, through encapsulation in particulate vectors, making structural changes that help binding to the presenting molecule CD1d, or injecting agonists covalently attached to recombinant CD1d. Variations on these same approaches are being used to enhance the APC-licencing function of iNKT cells in vivo to induce adaptive immune responses to associated tumour antigens. Looking ahead, a unique capacity of in vivo-activated iNKT cells to facilitate formation of resident memory CD8+ T cells is a new observation that could find a role in cancer therapy.
Collapse
Affiliation(s)
- Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
| | - Olivia K Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Ian F Hermans
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand; Malaghan Institute of Medical Research, Wellington, New Zealand.
| |
Collapse
|
47
|
Driver JP, de Carvalho Madrid DM, Gu W, Artiaga BL, Richt JA. Modulation of Immune Responses to Influenza A Virus Vaccines by Natural Killer T Cells. Front Immunol 2020; 11:2172. [PMID: 33193296 PMCID: PMC7606973 DOI: 10.3389/fimmu.2020.02172] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/10/2020] [Indexed: 12/20/2022] Open
Abstract
Influenza A viruses (IAVs) circulate widely among different mammalian and avian hosts and sometimes give rise to zoonotic infections. Vaccination is a mainstay of IAV prevention and control. However, the efficacy of IAV vaccines is often suboptimal because of insufficient cross-protection among different IAV genotypes and subtypes as well as the inability to keep up with the rapid molecular evolution of IAV strains. Much attention is focused on improving IAV vaccine efficiency using adjuvants, which are substances that can modulate and enhance immune responses to co-administered antigens. The current review is focused on a non-traditional approach of adjuvanting IAV vaccines by therapeutically targeting the immunomodulatory functions of a rare population of innate-like T lymphocytes called invariant natural killer T (iNKT) cells. These cells bridge the innate and adaptive immune systems and are capable of stimulating a wide array of immune cells that enhance vaccine-mediated immune responses. Here we discuss the factors that influence the adjuvant effects of iNKT cells for influenza vaccines as well as the obstacles that must be overcome before this novel adjuvant approach can be considered for human or veterinary use.
Collapse
Affiliation(s)
- John P Driver
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | | | - Weihong Gu
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Bianca L Artiaga
- Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Jürgen A Richt
- Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
48
|
Huang J, Zhou J, Ghinnagow R, Seki T, Iketani S, Soulard D, Paczkowski P, Tsuji Y, MacKay S, Cruz LJ, Trottein F, Tsuji M. Targeted Co-delivery of Tumor Antigen and α-Galactosylceramide to CD141 + Dendritic Cells Induces a Potent Tumor Antigen-Specific Human CD8 + T Cell Response in Human Immune System Mice. Front Immunol 2020; 11:2043. [PMID: 32973811 PMCID: PMC7461784 DOI: 10.3389/fimmu.2020.02043] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/27/2020] [Indexed: 02/01/2023] Open
Abstract
Active co-delivery of tumor antigens (Ag) and α-galactosylceramide (α-GalCer), a potent agonist for invariant Natural Killer T (iNKT) cells, to cross-priming CD8α+ dendritic cells (DCs) was previously shown to promote strong anti-tumor responses in mice. Here, we designed a nanoparticle-based vaccine able to target human CD141+ (BDCA3+) DCs - the equivalent of murine CD8α+ DCs – and deliver both tumor Ag (Melan A) and α-GalCer. This nanovaccine was inoculated into humanized mice that mimic the human immune system (HIS) and possess functional iNKT cells and CD8+ T cells, called HIS-CD8/NKT mice. We found that multiple immunizations of HIS-CD8/NKT mice with the nanovaccine resulted in the activation and/or expansion of human CD141+ DCs and iNKT cells and ultimately elicited a potent Melan-A-specific CD8+ T cell response, as determined by tetramer staining and ELISpot assay. Single-cell proteomics further detailed the highly polyfunctional CD8+ T cells induced by the nanovaccine and revealed their predictive potential for vaccine potency. This finding demonstrates for the first time the unique ability of human iNKT cells to license cross-priming DCs in vivo and adds a new dimension to the current strategy of cancer vaccine development.
Collapse
Affiliation(s)
- Jing Huang
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY, United States.,Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Jing Zhou
- IsoPlexis, Branford, CT, United States
| | - Reem Ghinnagow
- Centre d'Infection et d'Immunité de Lille, Inserm U1019, CNRS UMR 8204, CHU Lille, Institut Pasteur de Lille, University of Lille, Lille, France
| | - Toshiyuki Seki
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY, United States.,Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Sho Iketani
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY, United States.,Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, United States
| | - Daphnée Soulard
- Centre d'Infection et d'Immunité de Lille, Inserm U1019, CNRS UMR 8204, CHU Lille, Institut Pasteur de Lille, University of Lille, Lille, France
| | | | - Yukiko Tsuji
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY, United States
| | | | - Luis Javier Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - François Trottein
- Centre d'Infection et d'Immunité de Lille, Inserm U1019, CNRS UMR 8204, CHU Lille, Institut Pasteur de Lille, University of Lille, Lille, France
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY, United States.,Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
49
|
Ioannidis M, Cerundolo V, Salio M. The Immune Modulating Properties of Mucosal-Associated Invariant T Cells. Front Immunol 2020; 11:1556. [PMID: 32903532 PMCID: PMC7438542 DOI: 10.3389/fimmu.2020.01556] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/12/2020] [Indexed: 12/12/2022] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are unconventional T lymphocytes that express a semi-invariant T cell receptor (TCR) recognizing microbial vitamin B metabolites presented by the highly conserved major histocompatibility complex (MHC) class I like molecule, MR1. The vitamin B metabolites are produced by several commensal and pathogenic bacteria and yeast, but not viruses. Nevertheless, viral infections can trigger MAIT cell activation in a TCR-independent manner, through the release of pro-inflammatory cytokines by antigen-presenting cells (APCs). MAIT cells belong to the innate like T family of cells with a memory phenotype, which allows them to rapidly release Interferon (IFN)-γ, tumor necrosis factor (TNF)-α, and in some circumstances Interleukin (IL)-17 and IL-10, exerting an immunomodulatory role on the ensuing immune response, akin to iNKT cells and γδ T cells. Recent studies implicate MAIT cells in a variety of inflammatory, autoimmune diseases, and in cancer. In addition, through the analysis of the transcriptome of MAIT cells activated in different experimental conditions, an important function in tissue repair and control of immune homeostasis has emerged, shared with other innate-like T cells. In this review, we discuss these recent findings, focussing on the understanding of the molecular mechanisms underpinning MAIT cell activation and effector function in health and disease, which ultimately will aid in clinically harnessing this unique, not donor-restricted cell subtype.
Collapse
Affiliation(s)
- Melina Ioannidis
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Vincenzo Cerundolo
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Mariolina Salio
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
50
|
Ngiow SF, Young A. Re-education of the Tumor Microenvironment With Targeted Therapies and Immunotherapies. Front Immunol 2020; 11:1633. [PMID: 32849557 PMCID: PMC7399169 DOI: 10.3389/fimmu.2020.01633] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
The clinical success of cancer immunotherapies targeting PD-1 and CTLA-4 has ignited a substantial research effort to improve our understanding of tumor immunity. Recent studies have revealed that the immune contexture of a tumor influences therapeutic response and survival benefit for cancer patients. Identifying treatment modalities that limit immunosuppression, relieve T cell exhaustion, and potentiate effector functions in the tumor microenvironment (TME) is of much interest. In particular, combinatorial therapeutic approaches that re-educate the TME by limiting the accumulation of immunosuppressive immune cells, such as Foxp3 regulatory T cells (Tregs) and tumor-associated macrophages (TAMs), while promoting CD8+ and CD4+ effector T cell activity is critical. Here, we review key approaches to target these immunosuppressive immune cell subsets and signaling molecules and define the impact of these changes to the tumor milieu. We will highlight the preclinical and clinical evidence for their ability to improve anti-tumor immune responses as well as strategies and challenges for their implementation. Together, this review will provide understanding of therapeutic approaches to efficiently shape the TME and reinvigorate the immune response against cancer.
Collapse
Affiliation(s)
- Shin Foong Ngiow
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Arabella Young
- Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|