1
|
Metelitsa LS. A milestone method to make natural killer T cells. Nat Biotechnol 2025; 43:302-303. [PMID: 38744945 DOI: 10.1038/s41587-024-02243-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Affiliation(s)
- Leonid S Metelitsa
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer and Hematology Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
2
|
Boonchalermvichian C, Yan H, Gupta B, Rubin A, Baker J, Negrin RS. invariant Natural Killer T cell therapy as a novel therapeutic approach in hematological malignancies. FRONTIERS IN TRANSPLANTATION 2024; 3:1353803. [PMID: 38993780 PMCID: PMC11235242 DOI: 10.3389/frtra.2024.1353803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/04/2024] [Indexed: 07/13/2024]
Abstract
Invariant Natural Killer T cell therapy is an emerging platform of immunotherapy for cancer treatment. This unique cell population is a promising candidate for cell therapy for cancer treatment because of its inherent cytotoxicity against CD1d positive cancers as well as its ability to induce host CD8 T cell cross priming. Substantial evidence supports that iNKT cells can modulate myelomonocytic populations in the tumor microenvironment to ameliorate immune dysregulation to antagonize tumor progression. iNKT cells can also protect from graft-versus-host disease (GVHD) through several mechanisms, including the expansion of regulatory T cells (Treg). Ultimately, iNKT cell-based therapy can retain antitumor activity while providing protection against GVHD simultaneously. Therefore, these biological properties render iNKT cells as a promising "off-the-shelf" therapy for diverse hematological malignancies and possible solid tumors. Further the introduction of a chimeric antigen recetor (CAR) can further target iNKT cells and enhance function. We foresee that improved vector design and other strategies such as combinatorial treatments with small molecules or immune checkpoint inhibitors could improve CAR iNKT in vivo persistence, functionality and leverage anti-tumor activity along with the abatement of iNKT cell dysfunction or exhaustion.
Collapse
|
3
|
Moraes Ribeiro E, Secker KA, Nitulescu AM, Schairer R, Keppeler H, Wesle A, Schmid H, Schmitt A, Neuber B, Chmiest D, Podavini S, Märklin M, Klimovich B, Schmitt M, Korkmaz F, Lengerke C, Schneidawind C, Schneidawind D. PD-1 checkpoint inhibition enhances the antilymphoma activity of CD19-CAR-iNKT cells that retain their ability to prevent alloreactivity. J Immunother Cancer 2024; 12:e007829. [PMID: 38296597 PMCID: PMC10831439 DOI: 10.1136/jitc-2023-007829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2023] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Relapse and graft-versus-host disease (GVHD) are the main causes of death after allogeneic hematopoietic cell transplantation (HCT). Preclinical murine models and clinical data suggest that invariant natural killer T (iNKT) cells prevent acute and chronic GVHD. In addition, iNKT cells are crucial for efficient immune responses against malignancies and contribute to reduced relapse rates after transplantation. Chimeric antigen receptors (CAR) redirect effector cells to cell surface antigens and enhance killing of target cells. With this study, we aimed to combine enhanced cytotoxicity of CD19-CAR-iNKT cells against lymphoma cells with their tolerogenic properties. METHODS iNKT cells were isolated from peripheral blood mononuclear cells and transduced with an anti-CD19-CAR retrovirus. After in vitro expansion, the functionality of CD19-CAR-iNKT cells was assessed by flow cytometry, image stream analysis and multiplex analysis in single-stimulation or repeated-stimulation assays. Moreover, the immunoregulatory properties of CD19-CAR-iNKT cells were analyzed in apoptosis assays and in mixed lymphocyte reactions. The effect of checkpoint inhibition through nivolumab was analyzed in these settings. RESULTS In this study, we could show that the cytotoxicity of CD19-CAR-iNKT cells was mediated either through engagement of their CAR or their invariant T-cell receptor, which may circumvent loss of response through antigen escape. However, encounter of CD19-CAR-iNKT cells with their target induced a phenotype of exhaustion. Consequently, checkpoint inhibition increased cytokine release, cytotoxicity and survival of CD19-CAR-iNKT cells. Additionally, they showed robust suppression of alloreactive immune responses. CONCLUSION In this work, we demonstrate that CAR-iNKT cells are a powerful cytotherapeutic option to prevent or treat relapse while potentially reducing the risk of GVHD after allogeneic HCT.
Collapse
Affiliation(s)
- Emmanuelle Moraes Ribeiro
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Kathy-Ann Secker
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Ana-Maria Nitulescu
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Rebekka Schairer
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Hildegard Keppeler
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Anton Wesle
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Hannes Schmid
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Anita Schmitt
- Department of Oncology, Hematology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Oncology, Hematology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniela Chmiest
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland
| | - Silvia Podavini
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland
| | - Melanie Märklin
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Boris Klimovich
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Michael Schmitt
- Department of Oncology, Hematology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Fulya Korkmaz
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Claudia Lengerke
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Corina Schneidawind
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Dominik Schneidawind
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| |
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
|
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
|
6
|
Wang Y, Huang R, Wang Z, Xiong J, Wang X, Zhang X. Facing challenges with hope: universal immune cells for hematologic malignancies. Cancer Biol Med 2023; 20:229-247. [PMID: 37144558 PMCID: PMC10157807 DOI: 10.20892/j.issn.2095-3941.2022.0759] [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: 12/20/2022] [Accepted: 03/07/2023] [Indexed: 05/06/2023] Open
Abstract
Many patients have achieved a favorable overall survival rate since allogenic hematopoietic stem cell transplantation (allo-HSCT) has been widely implemented to treat hematologic malignancies. However, graft-versus-host disease (GVHD) and complications of immunosuppressive drugs after allo-HSCT are the main causes of non-relapse mortality and a poor quality of life. In addition, GVHD and infusion-induced toxicity still occur with donor lymphocyte infusions (DLIs) and chimeric antigen receptor (CAR) T-cell therapy. Because of the special immune tolerance characteristics and anti-tumor ability of universal immune cells, universal immune cell therapy may strongly reduce GVHD, while simultaneously reducing tumor burden. Nevertheless, widespread application of universal immune cell therapy is mainly restricted by poor expansion and persistence efficacy. Many strategies have been applied to improve universal immune cell proliferation and persistence efficacy, including the use of universal cell lines, signaling regulation and CAR technology. In this review we have summarized current advances in universal immune cell therapy for hematologic malignancies with a discussion of future perspectives.
Collapse
Affiliation(s)
- Yuqing Wang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing 400037, China
- Jinfeng Laboratory, Chongqing 400037, China
| | - Ruihao Huang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing 400037, China
| | - Zheng Wang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing 400037, China
| | - Jingkang Xiong
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing 400037, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing 400037, China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing 400037, China
- Jinfeng Laboratory, Chongqing 400037, China
| |
Collapse
|
7
|
Qin Y, Bao X, Zheng M. CD8 + T-cell immunity orchestrated by iNKT cells. Front Immunol 2023; 13:1109347. [PMID: 36741397 PMCID: PMC9889858 DOI: 10.3389/fimmu.2022.1109347] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/30/2022] [Indexed: 01/19/2023] Open
Abstract
CD8+ T cells belonging to the adaptive immune system play key roles in defending against viral infections and cancers. The current CD8+ T cell-based immunotherapy has emerged as a superior therapeutic avenue for the eradication of tumor cells and long-term prevention of their recurrence in hematologic malignancies. It is believed that an effective adaptive immune response critically relies on the help of the innate compartment. Invariant natural killer T (iNKT) cells are innate-like T lymphocytes that have been considered some of the first cells to respond to infections and can secrete a large amount of diverse cytokines and chemokines to widely modulate the innate and adaptive immune responders. Like CD8+ T cells, iNKT cells also play an important role in defense against intracellular pathogenic infections and cancers. In this review, we will discuss the CD8+ T-cell immunity contributed by iNKT cells, including iNKT cell-mediated cross-priming and memory formation, and discuss recent advances in our understanding of the mechanisms underlying memory CD8+ T-cell differentiation, as well as aging-induced impairment of T-cell immunity.
Collapse
|
8
|
Varghese B, Lynch L, Vriend LE, Draganov D, Clark JM, Kissick HT, Varghese S, Sanda MG, Dranoff G, Arredouani MS, Balk SP, Exley MA. Invariant NKT cell-augmented GM-CSF-secreting tumor vaccine is effective in advanced prostate cancer model. Cancer Immunol Immunother 2022; 71:2943-2955. [PMID: 35523889 PMCID: PMC10992623 DOI: 10.1007/s00262-022-03210-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
Invariant natural killer T cells (iNKT cells) express a semi-invariant T cell receptor that recognizes certain glycolipids (including α-galactosylceramide, αGC) bound to CD1d, and can induce potent antitumor responses. Here, we assessed whether αGC could enhance the efficacy of a GM-CSF-producing tumor cell vaccine in the transgenic SV40 T antigen-driven TRAMP prostate cancer model. In healthy mice, we initially found that optimal T cell responses were obtained with αGC-pulsed TRAMP-C2 cells secreting GM-CSF and milk fat globule epidermal growth factor protein-8 (MFG-E8) with an RGD to RGE mutation (GM-CSF/RGE TRAMP-C2), combined with systemic low dose IL-12. In a therapeutic model, transgenic TRAMP mice were then castrated at ~ 20 weeks, followed by treatment with the combination vaccine. Untreated mice succumbed to tumor by ~ 40 weeks, but survival was markedly prolonged by vaccine treatment, with most mice surviving past 80 weeks. Prostates in the treated mice were heavily infiltrated with T cells and iNKT cells, which both secreted IFNγ in response to tumor cells. The vaccine was not effective if the αGC, IL-12, or GM-CSF secretion was eliminated. Finally, immunized mice were fully resistant to challenge with TRAMP-C2 cells. Together these findings support further development of therapeutic vaccines that exploit iNKT cell activation.
Collapse
Affiliation(s)
- Bindu Varghese
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Sana Biotechnology Inc., Boston, MA, USA
| | - Lydia Lynch
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Brigham and Women's Hospital, 75 Francis St., NRB 6, Boston, MA, 02115, USA
| | - Lianne E Vriend
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Dobrin Draganov
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Sanofi Inc., San Diego, CA, USA
| | - Justice M Clark
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Haydn T Kissick
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Emory University, Atlanta, GA, USA
| | - Sharlin Varghese
- Medical Center School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Martin G Sanda
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Emory University, Atlanta, GA, USA
| | - Glenn Dranoff
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Novartis Biomedical Institutes of Research, Cambridge, MA, USA
| | - M Simo Arredouani
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Intellia Inc., Cambridge, MA, USA
| | - Steven P Balk
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.
| | - Mark A Exley
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.
- Brigham and Women's Hospital, 75 Francis St., NRB 6, Boston, MA, 02115, USA.
- Imvax Inc., Philadelphia, PA, USA.
- University of Manchester, Manchester, UK.
- MiNK Therapeutics Inc., New York, NY, USA.
| |
Collapse
|
9
|
Clarkson BDS, Johnson RK, Bingel C, Lothaller C, Howe CL. Preservation of antigen-specific responses in cryopreserved CD4 + and CD8 + T cells expanded with IL-2 and IL-7. J Transl Autoimmun 2022; 5:100173. [PMID: 36467614 PMCID: PMC9713293 DOI: 10.1016/j.jtauto.2022.100173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/31/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022] Open
Abstract
Objectives We sought to develop medium throughput standard operating procedures for screening cryopreserved human peripheral blood mononuclear cells (PBMCs) for CD4+ and CD8+ T cell responses to potential autoantigens. Methods Dendritic cells were loaded with a peptide cocktail from ubiquitous viruses or full-length viral protein antigens and cocultured with autologous T cells. We measured expression of surface activation markers on T cells by flow cytometry and cytometry by time of flight 24-72 h later. We tested responses among T cells freshly isolated from healthy control PBMCs, cryopreserved T cells, and T cells derived from a variety of T cell expansion protocols. We also compared the transcriptional profile of CD8+ T cells rested with interleukin (IL)7 for 48 h after 1) initial thawing, 2) expansion, and 3) secondary cryopreservation/thawing of expanded cells. To generate competent antigen presenting cells from PBMCs, we promoted differentiation of PBMCs into dendritic cells with granulocyte macrophage colony stimulating factor and IL-4. Results We observed robust dendritic cell differentiation from human PBMCs treated with 50 ng/mL GM-CSF and 20 ng/mL IL-4 in as little as 3 days. Dendritic cell purity was substantially increased by magnetically enriching for CD14+ monocytes prior to differentiation. We also measured antigen-dependent T cell activation in DC-T cell cocultures. However, polyclonal expansion of T cells with anti-CD3/antiCD28 abolished antigen-dependent upregulation of CD69 in our assay despite minimal transcriptional differences between rested CD8+ T cells before and after expansion. Furthermore, resting these expanded T cells in IL-2, IL-7 or IL-15 did not restore the antigen dependent responses. In contrast, T cells that were initially expanded with IL-2 + IL-7 rather than plate bound anti-CD3 + anti-CD28 retained responsiveness to antigen stimulation and these responses strongly correlated with responses measured at initial thawing. Significance While screening techniques for potential pathological autoantibodies have come a long way, comparable full-length protein target assays for screening patient T cells at medium throughput are noticeably lacking due to technical hurdles. Here we advance techniques that should have broad applicability to translational studies investigating cell mediated immunity in infectious or autoimmune diseases. Future studies are aimed at investigating possible CD8+ T cell autoantigens in MS and other CNS autoimmune diseases.
Collapse
Affiliation(s)
- Benjamin DS. Clarkson
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA,Corresponding author. Mayo Clinic, Guggenheim 1521C, 200 First Street SW, Rochester, MN, 55905.
| | | | - Corinna Bingel
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center, Heidelberg, Germany
| | | | - Charles L. Howe
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Division of Experimental Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
10
|
Characterization of a Glycolipid Synthase Producing α-Galactosylceramide in Bacteroides fragilis. Int J Mol Sci 2022; 23:ijms232213975. [PMID: 36430454 PMCID: PMC9692976 DOI: 10.3390/ijms232213975] [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: 10/18/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Glycolipids are complex molecules involved in important cellular processes. Among them, the glycosphingolipid α-galactosylceramide has proven to be of interest in biomedicine for its immunostimulatory capabilities. Given its structural requirements, the use of ceramide glycosyltransferase enzymes capable of synthesizing this molecule under in vivo or in vitro conditions is a potential production strategy. Several GT4 enzymes from Bacteroides fragilis were considered as potential candidates in addition to the known BF9343_3149, but only this one showed glycolipid synthase activity. The enzyme was expressed as a SUMO fusion protein to produce soluble protein. It is a non-processive glycosyltransferase that prefers UDP-Gal over UDP-Glc as a donor substrate, and maximum activity was found at pH 7.3 and around 30-35 °C. It does not require metal cations for activity as other GT4 enzymes, but Zn2+ inactivates the enzyme. The reaction occurs when the ceramide lipid acceptor is solubilized with BSA (100% conversion) but not when it is presented in mixed micelles, and anionic lipids do not increase activity, as in other membrane-associated glycolipid synthases. Further protein engineering to increase stability and activity can make feasible the enzymatic synthesis of α-GalCer for biomedical applications.
Collapse
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
Shimizu K, Ueda S, Kawamura M, Satoh M, Fujii SI. A single immunization with cellular vaccine confers dual protection against SARS-CoV-2 and cancer. Cancer Sci 2022; 113:2536-2547. [PMID: 35598170 PMCID: PMC9348309 DOI: 10.1111/cas.15434] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 11/29/2022] Open
Abstract
The efficacy of current coronavirus disease 2019 (COVID‐19) vaccines has been demonstrated; however, emerging evidence suggests insufficient protection in certain immunocompromised cancer patients. We previously developed a cell‐based anti‐cancer vaccine platform involving artificial adjuvant vector cells (aAVCs) capable of inducing a strong adaptive response by enhancing the innate immunity. aAVCs are target antigen‐transfected allogenic cells that simultaneously express the natural killer T‐cell ligand–CD1d complex on their surface. In the present study, we applied this system for targeting the severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) spike protein (CoV‐2‐S) using CoV‐2‐S‐expressing aAVCs (aAVC‐CoV‐2) and evaluated the immune response in a murine model. A single dose of aAVC‐CoV‐2 induced a large amount of CoV‐2‐S‐specific, multifunctional CTLs in addition to CD4+ T‐cell‐dependent anti‐CoV‐2‐S‐specific Abs. CoV‐2‐S‐specific CTLs infiltrated the lung parenchyma and persisted as long‐term memory T cells. Furthermore, we immunized mice with CoV‐2‐S‐ and tumor‐associated antigen (TAA)‐co‐expressing aAVCs (aAVC‐TAA/CoV‐2) and evaluated whether the anti‐SARS‐CoV‐2 and antitumor CTLs were elicited. We found that the aAVC‐TAA/CoV‐2‐S therapy exerted apparent antitumor effects and induced CoV‐2‐S‐specific CTLs. These findings suggest aAVC‐TAA/CoV‐2‐S therapy as a promising vaccine candidate for preventing COVID‐19, as well as enhancing the effectiveness of cancer therapies.
Collapse
Affiliation(s)
- Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Science, Yokohama, Kanagawa, Japan
| | - Shogo Ueda
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Science, Yokohama, Kanagawa, Japan
| | - Masami Kawamura
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Science, Yokohama, Kanagawa, Japan
| | - Mikiko Satoh
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Science, Yokohama, Kanagawa, Japan
| | - Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Science, Yokohama, Kanagawa, Japan.,Program for Drug Discovery and Medical Technology Platforms, RIKEN, Yokohama, Kanagawa, Japan
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
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
|
18
|
Azuma M, Ebihara T, Oshiumi H, Matsumoto M, Seya T. Cross-priming for antitumor CTL induced by soluble Ag + polyI:C depends on the TICAM-1 pathway in mouse CD11c(+)/CD8α(+) dendritic cells. Oncoimmunology 2021; 1:581-592. [PMID: 22934250 PMCID: PMC3429562 DOI: 10.4161/onci.19893] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PolyI:C is a nucleotide pattern molecule that induces cross-presentation of foreign Ag in myeloid dendritic cells (DC) and MHC Class I-dependent proliferation of cytotoxic T lymphocytes (CTL). DC (BM or spleen CD8α+) have sensors for dsRNA including polyI:C to signal facilitating cross-presentation. Endosomal TLR3 and cytoplasmic RIG-I/MDA5 are reportedly responsible for polyI:C sensing and presumed to deliver signal for cross-presentation via TICAM-1 (TRIF) and IPS-1 (MAVS, Cardif, VISA) adaptors, respectively. In fact, when tumor-associated Ag (TAA) was simultaneously taken up with polyI:C in DC, the DC cross-primed CTL specific to the TAA in a syngenic mouse model. Here we tested which of the TICAM-1 or IPS-1 pathway participate in cross-presentation of tumor-associated soluble Ag and retardation of tumor growth in the setting with a syngeneic tumor implant system, EG7/C57BL6, and exogenously challenged soluble Ag (EG7 lysate) and polyI:C. When EG7 lysate and polyI:C were subcutaneously injected in tumor-bearing mice, EG7 tumor growth retardation was observed in wild-type and to a lesser extent IPS-1−/− mice, but not TICAM-1−/− mice. IRF-3/7 were essential but IPS-1 and type I IFN were minimally involved in the polyI:C-mediated CTL proliferation. Although both TICAM-1 and IPS-1 contributed to CD86/CD40 upregulation in CD8α+ DC, H2Kb-SL8 tetramer and OT-1 proliferation assays indicated that OVA-recognizing CD8 T cells predominantly proliferated in vivo through TICAM-1 and CD8α+ DC is crucial in ex vivo analysis. Ultimately, tumor regresses > 8 d post polyI:C administration. The results infer that soluble tumor Ag induces tumor growth retardation, i.e., therapeutic potential, if the TICAM-1 signal coincidentally occurs in CD8α+ DC around the tumor.
Collapse
Affiliation(s)
- Masahiro Azuma
- Department of Microbiology and Immunology; Hokkaido University Graduate School of Medicine; Sapporo, Japan
| | | | | | | | | |
Collapse
|
19
|
Trujillo-Ocampo A, Cho HW, Clowers M, Pareek S, Ruiz-Vazquez W, Lee SE, Im JS. IL-7 During Antigenic Stimulation Using Allogeneic Dendritic Cells Promotes Expansion of CD45RA -CD62L +CD4 + Invariant NKT Cells With Th-2 Biased Cytokine Production Profile. Front Immunol 2020; 11:567406. [PMID: 33329531 PMCID: PMC7728799 DOI: 10.3389/fimmu.2020.567406] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/29/2020] [Indexed: 11/18/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are innate-like T lymphocytes cells that recognize glycolipid antigens associated with CD1d, non-classical antigen presenting proteins. They can drive either pro-inflammatory (Th-1) or anti-inflammatory (Th-2) immune microenvironment through the production of both Th-1 and Th-2 type cytokines upon activation, thus play a vital role in cancer, infection, and autoimmune diseases. Adoptive cell therapy using ex vivo expanded iNKT cells is a promising approach to enhance anti-tumor immunity or immunosuppression. However, overcoming phenotypic and functional heterogeneity and promoting in vivo persistency of iNKT cells remains to be a challenge. Here, we compared various methods for ex vivo expansion of human iNKT cells and assessed the quality of expansion, phenotype, and cytokine production profile of expanded iNKT cells. While a direct stimulation of iNKT cells in peripheral blood mononuclear cells with agonist glycolipid led to the expansion of iNKT cells in varying degrees, stimulation of enriched iNKT cells by irradiated autologous peripheral blood mononuclear cells or allogeneic dendritic cells resulted in consistent expansion of highly pure iNKT cells. Interestingly, the mode of antigenic stimulation influenced the dominant subtype of expanded iNKT cells. Further, we evaluated whether additional IL-7 or IL-15 during antigenic stimulation with allogeneic dendritic cells can improve the phenotypic heterogeneity and modify cytokine production profile of iNKT cells expanded from 18 consecutive donors. The presence of IL-7 or IL-15 during antigenic stimulation did not affect the fold of expansion or purity of expanded iNKT cells. However, IL-7, but not IL-15, led to a better expansion of CD4+ iNKT cells, enhanced Th-2 type cytokine production of CD4+ iNKT cells, and maintained the expansion of central memory (CD45RA-CD62L+) CD4+ iNKT cells. Our results suggest the addition of IL-7 during antigenic stimulation with allogeneic dendritic cells can promote the expansion of CD62L+Th-2+CD4+ human iNKT cells that can be used as novel immunotherapeutic to control excessive inflammation to treat various autoimmune diseases.
Collapse
Affiliation(s)
- Abel Trujillo-Ocampo
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hyun-Woo Cho
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael Clowers
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sumedha Pareek
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Wilfredo Ruiz-Vazquez
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sung-Eun Lee
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jin S Im
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
20
|
Grasso C, Field CS, Tang CW, Ferguson PM, J Compton B, Anderson RJ, Painter GF, Weinkove R, F Hermans I, Berridge MV. Vaccines adjuvanted with an NKT cell agonist induce effective T-cell responses in models of CNS lymphoma. Immunotherapy 2020; 12:395-406. [PMID: 32316797 DOI: 10.2217/imt-2019-0134] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The efficacy of anti-lymphoma vaccines that exploit the cellular adjuvant properties of activated natural killer T (NKT) cells were examined in mouse models of CNS lymphoma. Materials & methods: Vaccines were prepared by either loading the NKT cell agonist, α-galactosylceramide onto irradiated and heat-shocked B- and T-lymphoma cells, or chemically conjugating α-galactosylceramide to MHC-binding peptides from a lymphoma-associated antigen. Vaccine efficacy was analyzed in mice bearing intracranial tumors. Results: Both forms of vaccine proved to be effective in preventing lymphoma engraftment through activity of T cells that accessed the CNS. Established lymphoma was harder to treat with responses constrained by Tregs, but this could be overcome by depleting Tregs prior to vaccination. Conclusion: Simply designed NKT cell-activating vaccines enhance T-cell responses and have the potential to protect against CNS lymphoma development or prevent CNS relapse. To be effective against established CNS lymphoma, vaccines need to be combined with Treg suppression.
Collapse
Affiliation(s)
- Carole Grasso
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6242, New Zealand
| | - Cameron S Field
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6242, New Zealand
| | - Ching-Wen Tang
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6242, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Private Bag 92019, Auckland 1042, New Zealand
| | - Peter M Ferguson
- Melanoma Institute Australia, 40 Rocklands Road, Wollstonecraft, NSW 2065, Australia
| | - Benjamin J Compton
- The Ferrier Research Institute, Victoria University of Wellington, P.O. Box 33436, Lower Hutt 5046, New Zealand
| | - Regan J Anderson
- The Ferrier Research Institute, Victoria University of Wellington, P.O. Box 33436, Lower Hutt 5046, New Zealand
| | - Gavin F Painter
- The Ferrier Research Institute, Victoria University of Wellington, P.O. Box 33436, Lower Hutt 5046, New Zealand
| | - Robert Weinkove
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6242, New Zealand.,Wellington Blood & Cancer Centre, Capital & Coast District Health Board, P.O. Box 7902, Wellington 6242, New Zealand.,Department of Pathology & Molecular Medicine, University of Otago Wellington, P.O. Box 7343, Wellington 6242, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6242, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Private Bag 92019, Auckland 1042, New Zealand
| | - Michael V Berridge
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6242, New Zealand
| |
Collapse
|
21
|
Shimizu K, Iyoda T, Yamasaki S, Kadowaki N, Tojo A, Fujii SI. NK and NKT Cell-Mediated Immune Surveillance against Hematological Malignancies. Cancers (Basel) 2020; 12:cancers12040817. [PMID: 32231116 PMCID: PMC7226455 DOI: 10.3390/cancers12040817] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023] Open
Abstract
Recent cancer treatment modalities have been intensively focused on immunotherapy. The success of chimeric antigen receptor T cell therapy for treatment of refractory B cell acute lymphoblastic leukemia has pushed forward research on hematological malignancies. Among the effector types of innate lymphocytes, natural killer (NK) cells show great importance in immune surveillance against infectious and tumor diseases. Particularly, the role of NK cells has been argued in either elimination of target tumor cells or escape of tumor cells from immune surveillance. Therefore, an NK cell activation approach has been explored. Recent findings demonstrate that invariant natural killer T (iNKT) cells capable of producing IFN-γ when optimally activated can promptly trigger NK cells. Here, we review the role of NKT and/or NK cells and their interaction in anti-tumor responses by highlighting how innate immune cells recognize tumors, exert effector functions, and amplify adaptive immune responses. In addition, we discuss these innate lymphocytes in hematological disorders, particularly multiple myeloma and acute myeloid leukemia. The immune balance at different stages of both diseases is explored in light of disease progression. Various types of innate immunity-mediated therapeutic approaches, recent advances in clinical immunotherapies, and iNKT-mediated cancer immunotherapy as next-generation immunotherapy are then discussed.
Collapse
Affiliation(s)
- Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
- Correspondence: (K.S.); (S.-i.F.); Tel.: +81-45-503-7062 (K.S. & S.-i.F.); Fax: +81-45-503-7061 (K.S. & S.-i.F.)
| | - Tomonori Iyoda
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
| | - Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
| | - Norimitsu Kadowaki
- Department of Internal Medicine, Hematology, Rheumatology and Respiratory Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan;
| | - Arinobu Tojo
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Minato, Tokyo 108-8639, Japan;
| | - Shin-ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
- Correspondence: (K.S.); (S.-i.F.); Tel.: +81-45-503-7062 (K.S. & S.-i.F.); Fax: +81-45-503-7061 (K.S. & S.-i.F.)
| |
Collapse
|
22
|
Fu S, He K, Tian C, Sun H, Zhu C, Bai S, Liu J, Wu Q, Xie D, Yue T, Shen Z, Dai Q, Yu X, Zhu S, Liu G, Zhou R, Duan S, Tian Z, Xu T, Wang H, Bai L. Impaired lipid biosynthesis hinders anti-tumor efficacy of intratumoral iNKT cells. Nat Commun 2020; 11:438. [PMID: 31974378 PMCID: PMC6978340 DOI: 10.1038/s41467-020-14332-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022] Open
Abstract
Dysfunction of invariant natural killer T (iNKT) cells in tumor microenvironment hinders their anti-tumor efficacy, and the underlying mechanisms remain unclear. Here we report that iNKT cells increase lipid biosynthesis after activation, and that is promoted by PPARγ and PLZF synergically through enhancing transcription of Srebf1. Among those lipids, cholesterol is required for the optimal IFN-γ production from iNKT cells. Lactic acid in tumor microenvironment reduces expression of PPARγ in intratumoral iNKT cells and consequently diminishes their cholesterol synthesis and IFN-γ production. Importantly, PPARγ agonist pioglitazone, a thiazolidinedione drug for type 2 diabetes, successfully restores IFN-γ production in tumor-infiltrating iNKT cells from both human patients and mouse models. Combination of pioglitazone and alpha-galactosylceramide treatments significantly enhances iNKT cell-mediated anti-tumor immune responses and prolongs survival of tumor-bearing mice. Our studies provide a strategy to augment the anti-tumor efficacy of iNKT cell-based immunotherapies via promoting their lipid biosynthesis. Lipid metabolism has been linked to iNKT function largely as it impacts processing and presentation of lipids they recognize. Here the authors show that iNKT-intrinsic lipid biosynthesis is important for their function but is impaired in tumors, and its restoration with PPARγ agonist drugs promotes anti-tumor iNKT response.
Collapse
Affiliation(s)
- Sicheng Fu
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Kaixin He
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Chenxi Tian
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hua Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
| | - Chenwen Zhu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
| | - Shiyu Bai
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Jiwei Liu
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Qielan Wu
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Di Xie
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ting Yue
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Zhuxia Shen
- Department of Cardiology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, 200040, China
| | - Qingqing Dai
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiaojun Yu
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Shu Zhu
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Gang Liu
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, 230027, China
| | - Rongbin Zhou
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Shengzhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Zhigang Tian
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Tao Xu
- National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China.
| | - Li Bai
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China. .,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
23
|
Fujii SI, Shimizu K. Immune Networks and Therapeutic Targeting of iNKT Cells in Cancer. Trends Immunol 2019; 40:984-997. [PMID: 31676264 DOI: 10.1016/j.it.2019.09.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 01/08/2023]
Abstract
One of the primary goals in tumor immunotherapy is to reset the immune system from tolerogenic to immunogenic - a process in which invariant natural killer T (iNKT) cells are implicated. iNKT cells develop in the thymus and perform immunosurveillance against tumor cells peripherally. When optimally stimulated, iNKT cells differentiate and display more efficient immune functions. Some cells survive and act as effector memory cells. We discuss the putative roles of iNKT cells in antitumor immunity, and posit that it may be possible to develop novel therapeutic strategies to treat cancers using iNKT cells. In particular, we highlight the challenge of uniquely energizing iNKT cell-licensed dendritic cells to serve as effective immunoadjuvants for both arms of the immune system, thus coupling immunological networks.
Collapse
Affiliation(s)
- Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa, Japan.
| | - Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa, Japan
| |
Collapse
|
24
|
Shimizu K, Iyoda T, Okada M, Yamasaki S, Fujii SI. Immune suppression and reversal of the suppressive tumor microenvironment. Int Immunol 2019; 30:445-454. [PMID: 29939325 DOI: 10.1093/intimm/dxy042] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 06/22/2018] [Indexed: 12/18/2022] Open
Abstract
Most tumors employ multiple strategies to attenuate T-cell-mediated immune responses. In particular, immune suppression surrounding the tumor is achieved by interfering with antigen-presenting cells and effector T cells. Controlling both the tumor and the tumor microenvironment (TME) is critical for cancer treatment. Checkpoint blockade therapy can overcome tumor-induced immune suppression, but more than half of the patients fail to respond to this treatment; therefore, more effective cancer immunotherapies are needed. Generation of an anti-tumor immune response is a multi-step process of immune activation against the tumor that requires effector T cells to recognize and exert toxic effects against tumor cells, for which two strategies are employed-inhibition of various types of immune suppressor cells, such as myeloid cells and regulatory T cells, and establishment of anti-tumor immune surveillance including, activation of natural killer cells and cytotoxic T cells. It was recently shown that anti-cancer drugs not only directly kill tumor cells, but also influence the immune response to cancer by promoting immunogenic cell death, enhancing antigen presentation or depleting immunosuppressive cells. Herein, we review the mechanisms by which tumors exert immune suppression as well as their regulation. We then discuss how the complex reciprocal interactions between immunosuppressive and immunostimulatory cells influence immune cell dynamics in the TME. Finally, we highlight the new therapies that can reverse immune suppression in the TME and promote anti-tumor immunity.
Collapse
Affiliation(s)
- Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa, Japan
| | - Tomonori Iyoda
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa, Japan
| | - Masahiro Okada
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa, Japan
| | - Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa, Japan
| | - Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa, Japan
| |
Collapse
|
25
|
Paul S, Chhatar S, Mishra A, Lal G. Natural killer T cell activation increases iNOS +CD206 - M1 macrophage and controls the growth of solid tumor. J Immunother Cancer 2019; 7:208. [PMID: 31387637 PMCID: PMC6685184 DOI: 10.1186/s40425-019-0697-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/30/2019] [Indexed: 01/12/2023] Open
Abstract
Background NKT cells play an important role in anti-tumor immunity. Alpha-galactosylceramide (α-GalCer), a synthetic glycolipid is presented to natural killer T (NKT) cells by most antigen-presenting cells through CD1d molecules leading to activation of NKT cells. However, the precise mechanisms of how α-GalCer-activated NKT regulate the polarization of the macrophages and effector T cells in the solid tumor are not studied adequately. Methods We induced solid tumor in C57BL/6 mice by subcutaneous injection of B16F10 cell line (1 X 106 cells) and monitored the tumor growth. Animals were given an intraperitoneal injection of α-GalCer (2 μg/injection) in 200 μl PBS on day + 1, + 5, + 10, + 15, and + 20 (with respect to tumor cell injection). Immune cells were characterized using flow cytometry and immunofluorescence staining. NK cells, Gr1+ cells, and F4/80+ macrophages in the mice were depleted by intravenous injection of cell-specific antibodies. Statistical analysis was performed using Student’s t-test or one-way ANOVA. Results Our results showed that intratumoral NKT cells have a lower frequency of CD69, CD25, CD122, and IFN-γR expression; produced less inflammatory cytokines such as IFN-γ, TNF-α, and GM-CSF; higher frequency CD62L+ NKT cells; and also showed reduced proliferation as compared to the splenic NKT cells. Mice treated with α-GalCer showed a significantly increased frequency of IFN-γ-producing NKT cells, CD8+ T cells, and effector Th1 cells. Depletion of NK cells in α-GalCer-treated mice showed a lower frequency of IFN-γ-producing CD4+ and CD8+ T cells in the tumor and prevented the α-GalCer-induced tumor growth. NKT cell activation with α-GalCer treatment significantly increased the iNOS+CD206− M1-macrophages and reduced the iNOS−CD206+ M2-macrophages in the spleen and tumor, and depletion of F4/80+ macrophages prevented the α-GalCer-induced reduction in the tumor growth. Conclusions We showed that activation of NKT cell with α-GalCer modulates the frequency of M1-macrophages and effector Th1 cells in the secondary lymphoid tissues and tumor microenvironment and inhibit tumor growth. The finding suggests that activation of NKT cells with α-GalCer may provide an effective anti-cancer outcome. Electronic supplementary material The online version of this article (10.1186/s40425-019-0697-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sourav Paul
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, MH-411007, India
| | - Sushanta Chhatar
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, MH-411007, India
| | - Amrita Mishra
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, MH-411007, India
| | - Girdhari Lal
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, MH-411007, India.
| |
Collapse
|
26
|
Shimizu K, Yamasaki S, Sakurai M, Yumoto N, Ikeda M, Mishima-Tsumagari C, Kukimoto-Niino M, Watanabe T, Kawamura M, Shirouzu M, Fujii SI. Granzyme A Stimulates pDCs to Promote Adaptive Immunity via Induction of Type I IFN. Front Immunol 2019; 10:1450. [PMID: 31293597 PMCID: PMC6606709 DOI: 10.3389/fimmu.2019.01450] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 06/10/2019] [Indexed: 01/01/2023] Open
Abstract
Granzyme A (GzmA), together with perforin, are well-known for their cytotoxic activity against tumor or virus-infected cells. In addition to this cytotoxic function, GzmA stimulates several immune cell types and induces inflammation in the absence of perforin, however, its effect on the dendritic cell (DC) is unknown. In the current study, we showed that recombinant GzmA induced the phenotypic maturation of plasmacytoid DCs (pDCs) and conventional DCs (cDCs), but not their apoptosis. Particularly, GzmA made pDCs more functional, thus leading to production of type I interferon (IFN) via the TLR9-MyD88 pathway. We also demonstrated that GzmA binds TLR9 and co-localizes with it in endosomes. When co-administered with antigen, GzmA acted as a powerful adjuvant for eliciting antigen-specific cytotoxic CD8+ T lymphocytes (CTLs) that protected mice from tumor challenge. The induction of CTL was completely abolished in XCR1+ DC-depleted mice, whereas it was reduced to less than half in pDC-depleted or IFN-α/β receptor knockout mice. Thus, CTL cross-priming was dependent on XCR1+cDC and also type I IFN, which was produced by GzmA-activated pDCs. These results indicate that GzmA -stimulated pDCs enhance the cross-priming activity of cDCs in situ. We also showed that the adjuvant effect of GzmA is superior to CpG-ODN and LPS. Our findings highlight the ability of GzmA to bridge innate and adaptive immune responses via pDC help and suggest that GzmA may be useful as a vaccine adjuvant.
Collapse
Affiliation(s)
- Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Maki Sakurai
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Noriko Yumoto
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mariko Ikeda
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Chiemi Mishima-Tsumagari
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Mutsuko Kukimoto-Niino
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Takashi Watanabe
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masami Kawamura
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
27
|
Zhang Y, Springfield R, Chen S, Li X, Feng X, Moshirian R, Yang R, Yuan W. α-GalCer and iNKT Cell-Based Cancer Immunotherapy: Realizing the Therapeutic Potentials. Front Immunol 2019; 10:1126. [PMID: 31244823 PMCID: PMC6562299 DOI: 10.3389/fimmu.2019.01126] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/03/2019] [Indexed: 01/15/2023] Open
Abstract
NKT cells are CD1d-restricted innate-like T cells expressing both T cell receptor and NK cell markers. The major group of NKT cells in both human and mice is the invariant NKT (iNKT) cells and the best-known function of iNKT cells is their potent anti-tumor function in mice. Since its discovery 25 years ago, the prototype ligand of iNKT cells, α-galactosylceramide (α-GalCer) has been used in over 30 anti-tumor clinical trials with mostly suboptimal outcomes. To realize its therapeutic potential, numerous preclinical models have been developed to optimize the scheme and strategies for α-GalCer-based cancer immunotherapies. Nevertheless, since there is no standard protocol for α-GalCer delivery, we reviewed the preclinical studies with a focus on B16 melanoma model in the goal of identifying the best treatment schemes for α-GalCer treatment. We then reviewed the current progress in developing more clinically relevant mouse models for these preclinical studies, most notably the generation of new mouse models with a humanized CD1d/iNKT cell system. With ever-emerging novel iNKT cell ligands, invention of novel α-GalCer delivery strategies and significantly improved preclinical models for optimizing these new strategies, one can be hopeful that the full potential of anti-tumor potential for α-GalCer will be realized in the not too distant future.
Collapse
Affiliation(s)
- Yingting Zhang
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ryan Springfield
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Siyang Chen
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Xin Li
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Xiaotian Feng
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rosa Moshirian
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rirong Yang
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Weiming Yuan
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
28
|
A Characterization of Dendritic Cells and Their Role in Immunotherapy in Glioblastoma: From Preclinical Studies to Clinical Trials. Cancers (Basel) 2019; 11:cancers11040537. [PMID: 30991681 PMCID: PMC6521200 DOI: 10.3390/cancers11040537] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM) is the most common and fatal primary central nervous system malignancy in adults with a median survival of less than 15 months. Surgery, radiation, and chemotherapy are the standard of care and provide modest benefits in survival, but tumor recurrence is inevitable. The poor prognosis of GBM has made the development of novel therapies targeting GBM of paramount importance. Immunotherapy via dendritic cells (DCs) has garnered attention and research as a potential strategy to boost anti-tumor immunity in recent years. As the “professional” antigen processing and presenting cells, DCs play a key role in the initiation of anti-tumor immune responses. Pre-clinical studies in GBM have shown long-term tumor survival and immunological memory in murine models with stimulation of DC activity with various antigens and costimulatory molecules. Phase I and II clinical trials of DC vaccines in GBM have demonstrated some efficacy in improving the median overall survival with minimal to no toxicity with promising initial results from the first Phase III trial. However, there remains no standardization of vaccines in terms of which antigens are used to pulse DCs ex vivo, sites of DC injection, and optimal adjuvant therapies. Future work with DC vaccines aims to elucidate the efficacy of DC-based therapy alone or in combination with other immunotherapy adjuvants in additional Phase III trials.
Collapse
|
29
|
Escribà-Garcia L, Alvarez-Fernández C, Caballero AC, Schaub R, Sierra J, Briones J. The novel agonistic iNKT-cell antibody NKT14m induces a therapeutic antitumor response against B-cell lymphoma. Oncoimmunology 2019; 8:e1546543. [PMID: 30713807 DOI: 10.1080/2162402x.2018.1546543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/24/2018] [Accepted: 11/02/2018] [Indexed: 02/02/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are a small population of T lymphocytes that expresses an invariant T cell receptor with a unique specificity for glycolipid antigens. Their activation using the glycolipid α-galactosylceramide (α-GalCer) triggers innate and adaptive immune responses. The use of α-GalCer in preclinical models as a single antitumor treatment showed moderate effect, but its efficacy in cancer patients was less effective. In addition, this glycolipid induces long-term iNKT-cell anergy precluding the possibility of retreatment. Recently, the first murine iNKT-cell agonistic antibody, NKT14m, has been developed. Here, we analyzed, for the first time, the antitumor efficacy of NKT14m in a B-cell lymphoma model. In a therapeutic setting, a single dose of NKT14m had a moderate antitumor efficacy that was associated with an increase of IFN-γ producing iNKT cells even after a second dose of the NKT14m antibody. Importantly, the combination of a single dose of NKT14m with cyclophosphamide had a potent antitumor efficacy and long-lasting immunity in vivo. Our findings provide the first evidence of the in vivo antitumor efficacy of NKT14m antibody, showing that, either alone or in combination with chemotherapy, induces an effective antitumor response. These results open new opportunities for iNKT-cell mediated immunotherapy to treat B-cell lymphoma.
Collapse
Affiliation(s)
- Laura Escribà-Garcia
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Carmen Alvarez-Fernández
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Ana Carolina Caballero
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | | | - Jorge Sierra
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Autonomous University, Barcelona, Spain
| | - Javier Briones
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Autonomous University, Barcelona, Spain
| |
Collapse
|
30
|
Godfrey DI, Le Nours J, Andrews DM, Uldrich AP, Rossjohn J. Unconventional T Cell Targets for Cancer Immunotherapy. Immunity 2018; 48:453-473. [PMID: 29562195 DOI: 10.1016/j.immuni.2018.03.009] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 02/07/2023]
Abstract
Most studies on the immunotherapeutic potential of T cells have focused on CD8 and CD4 T cells that recognize peptide antigens (Ag) presented by polymorphic major histocompatibility complex (MHC) class I and MHC class II molecules, respectively. However, unconventional T cells, which interact with MHC class Ib and MHC-I like molecules, are also implicated in tumor immunity, although their role therein is unclear. These include unconventional T cells targeting MHC class Ib molecules such as HLA-E and its murine ortholog Qa-1b, natural killer T (NKT) cells, mucosal associated invariant T (MAIT) cells, and γδ T cells. Here, we review the current understanding of the roles of these unconventional T cells in tumor immunity and discuss why further studies into the immunotherapeutic potential of these cells is warranted.
Collapse
Affiliation(s)
- Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Jérôme Le Nours
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Daniel M Andrews
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Adam P Uldrich
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia; Institute of Infection and Immunity, Cardiff University, School of Medicine, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
31
|
Kerage D, Soon MSF, Doff BL, Kobayashi T, Nissen MD, Lam PY, Leggatt GR, Mattarollo SR. Therapeutic vaccination with 4-1BB co-stimulation eradicates mouse acute myeloid leukemia. Oncoimmunology 2018; 7:e1486952. [PMID: 30288351 DOI: 10.1080/2162402x.2018.1486952] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 12/17/2022] Open
Abstract
Immunomodulatory therapies can effectively control haematological malignancies. Previously we reported the effectiveness of combination immunotherapies that centre on 4-1BB-targeted co-stimulation of CD8 + T cells, particularly when simultaneously harnessing the immune adjuvant properties of Natural Killer T (NKT) cells. The objective of this study was to assess the effectiveness of agonistic anti-4-1BB antibody-based combination therapy against two aggressive forms of acute myeloid leukemia (AML). Anti-4-1BB treatment alone resulted in transient suppression of established AML-ETO9a tumor growth in 50% of mice, however the majority of these mice subsequently succumbed to disease. Combining alpha-galactosylceramide (α-GalCer)-loaded tumor cell vaccination with anti-4-1BB antibody treatment increased the proportion of responding mice to 100%, and protection led to long-term, tumor-free survival, demonstrating complete eradication of AML. This finding was extended to established mixed lymphocytic leukemia (MLL)-AF9 tumors, whereby vaccine plus anti-4-1BB combination similarly resulted in 100% protection. The addition of anti-PD-1 to anti-4-1BB treatment, although improving survival outcomes compared to anti-4-1BB alone, was not as effective as NKT cell vaccination. The effectiveness of 4-1BB combination therapies was dependent on IFN-γ signaling within host cells, but not tumors. Vaccine plus anti-4-1BB therapy elicited potent generation of functional effector and memory CD8 + T cells in all tumor-associated organs. Therapy induced KLRG1+ effector CD8 T cells were the most effective at controlling disease. We show that combining NKT cell-targeting vaccination with anti-4-1BB provides excellent therapeutic responses against AML and MLL in mice, and these results will guide ongoing efforts in finding immunotherapeutic solutions against acute myeloid leukemias.
Collapse
Affiliation(s)
- Daniel Kerage
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Megan S F Soon
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Brianna L Doff
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Takumi Kobayashi
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Michael D Nissen
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Pui Yeng Lam
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Graham R Leggatt
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Stephen R Mattarollo
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| |
Collapse
|
32
|
Abstract
Type I or invariant natural killer T cells belong to a unique lineage of innate T cells, which express markers of both T lymphocytes and NK cells, namely T cell receptor (TCR) and NK1.1 (CD161C), respectively. Thus, apart from direct killing of target cells like NK cells, and they also produce a myriad of cytokines which modulate the adaptive immune responses. Unlike traditional T cells which carry a conventional αβ TCR, NKT cells express semi-invariant TCR - Vα14-Jα18, coupled with Vβ8, Vβ7 and Vβ2 in mice. In humans, the invariant TCR is composed of Vα24-Jα18, coupled with Vβ11.
Collapse
Affiliation(s)
- Kalyani Pyaram
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, USA
| | - Viveka Nand Yadav
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, USA
| |
Collapse
|
33
|
Fujii SI, Yamasaki S, Sato Y, Shimizu K. Vaccine Designs Utilizing Invariant NKT-Licensed Antigen-Presenting Cells Provide NKT or T Cell Help for B Cell Responses. Front Immunol 2018; 9:1267. [PMID: 29915600 PMCID: PMC5995044 DOI: 10.3389/fimmu.2018.01267] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/22/2018] [Indexed: 12/26/2022] Open
Abstract
Vaccines against a variety of infectious diseases have been developed and tested. Although there have been some notable successes, most are less than optimal or have failed outright. There has been discussion about whether either B cells or dendritic cells (DCs) could be useful for the development of antimicrobial vaccines with the production of high titers of antibodies. Invariant (i)NKT cells have direct antimicrobial effects as well as adjuvant activity, and iNKT-stimulated antigen-presenting cells (APCs) can determine the form of the ensuing humoral and cellular immune responses. In fact, upon activation by ligand, iNKT cells can stimulate both B cells and DCs as via either cognate or non-cognate help. iNKT-licensed DCs generate antigen-specific follicular helper CD4+ T cells, which in turn stimulate B cells, thus leading to long-term antigen-specific antibody production. Follicular helper iNKT cell-licensed B cells generally produce rapid, but short-term antibody. However, under some conditions in the presence of Th cells, the antibody production can be prolonged. With regards to humoral immunity, the quality and quantity of Ab produced depends on the APC type and the form of the vaccine. In terms of cellular immunity and, in particular, the induction of cytotoxic CD8+ T cells, iNKT-licensed DCs show prominent activity. In this review, we discuss differences in iNKT-stimulated APC types and the quality of the ensuing immune response, and also discuss their application in vaccine models to develop successful preventive immunotherapy against infectious diseases.
Collapse
Affiliation(s)
- Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Yusuke Sato
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| |
Collapse
|
34
|
Okazaki S, Iwasaki T, Yuba E, Watarai S. Evaluation of pH-sensitive fusogenic polymer-modified liposomes co-loaded with antigen and α-galactosylceramide as an anti-tumor vaccine. J Vet Med Sci 2017; 80:197-204. [PMID: 29311431 PMCID: PMC5836753 DOI: 10.1292/jvms.17-0491] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
pH-Sensitive fusogenic polymer-modified (pH-sensitive) liposomes co-loaded with tumor
model antigen, ovalbumin (OVA), and adjuvant, α-galactosylceramide (α-GalCer) were
fabricated and administered subcutaneously into mice. The ability of pH-sensitive
liposomes containing OVA and α-GalCer to stimulate cellular and humoral immune responses
in vivo was compared with OVA-encapsulating pH-sensitive liposomes as
well as with OVA alone. After immunization, significant OVA-specific antibodies were
detected in the serum. When sera were analyzed for isotype distribution, antigen-specific
IgG1 antibody responses were noted in mice immunized with OVA alone, whereas immunization
with OVA-containing pH-sensitive liposomes and with pH-sensitive liposomes containing OVA
and α-GalCer resulted in the induction of OVA-specific IgG1 and IgG2b antibody responses.
Moreover, more substantial production of IFN-γ and IL-4 was demonstrated in spleen cells
from mice immunized with pH-sensitive liposomes having OVA and α-GalCer than
OVA-containing pH-sensitive liposomes in vitro. Spleen cells from the
immunized mice showed strong cytotoxic activity against E.G7-OVA tumor cells. In addition,
prophylactic vaccination efficacy against tumor formation was evaluated. In all mice
immunized with pH-sensitive liposomes having OVA and α-GalCer, immunization provided
substantial protection from tumor formation. The therapeutic efficacy of pH-sensitive
liposomes containing OVA and α-GalCer against already established E.G7-OVA tumors was also
investigated. Tumor growth was reduced significantly in all mice treated with pH-sensitive
liposomes having OVA and α-GalCer. The provided evidence on the advantage of antigen and
α-GalCer co-encapsulation into pH-sensitive liposomes should be considered in the design
of future cancer vaccines for prophylactic and therapeutic purposes.
Collapse
Affiliation(s)
- Seiji Okazaki
- Division of Veterinary Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Izumisano, Osaka 598-8531, Japan
| | - Tadashi Iwasaki
- Division of Veterinary Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Izumisano, Osaka 598-8531, Japan
| | - Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, Sakai, Osaka 599-8531, Japan
| | - Shinobu Watarai
- Division of Veterinary Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Izumisano, Osaka 598-8531, Japan
| |
Collapse
|
35
|
Teyton L. New Directions for Natural Killer T Cells in the Immunotherapy of Cancer. Front Immunol 2017; 8:1480. [PMID: 29209309 PMCID: PMC5701619 DOI: 10.3389/fimmu.2017.01480] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/23/2017] [Indexed: 01/07/2023] Open
Abstract
Natural killer T (NKT) cells have been placed at the interface between innate and adaptive immunity by a long series of experiments that convincingly showed that beyond cytokine secretion and NK cell recruitment, NKT cells were coordinating dendritic cell and B cell maturation through direct membrane contacts and initiate productive responses. As such, NKT cells are the cellular adjuvant of many immune reactions and have functions that go much beyond what their name encapsulates. In addition, the initial discovery of the ligands of NKT cells is deeply linked to cancer biology and therapy. However, for a host of reasons, animal models in which agonists of NKT cells were used did not translate well to human cancers. A systematic reassessment of NKT cells role in tumorigenesis, especially spontaneous one, is now accessible using single cell analysis technologies both in mouse and man, and should be taken advantage of. Similarly, the migration, localization, phenotype of NKT cells following induced expansion after injection of an agonist can be examined at the single cell level. This technological revolution will help evaluate where and how NKT cells can be used in cancer.
Collapse
Affiliation(s)
- Luc Teyton
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
36
|
Lam PY, Nissen MD, Mattarollo SR. Invariant Natural Killer T Cells in Immune Regulation of Blood Cancers: Harnessing Their Potential in Immunotherapies. Front Immunol 2017; 8:1355. [PMID: 29109728 PMCID: PMC5660073 DOI: 10.3389/fimmu.2017.01355] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/03/2017] [Indexed: 01/03/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are a unique innate T lymphocyte population that possess cytolytic properties and profound immunoregulatory activities. iNKT cells play an important role in the immune surveillance of blood cancers. They predominantly recognize glycolipid antigens presented on CD1d, but their activation and cytolytic activities are not confined to CD1d expressing cells. iNKT cell stimulation and subsequent production of immunomodulatory cytokines serve to enhance the overall antitumor immune response. Crucially, the activation of iNKT cells in cancer often precedes the activation and priming of other immune effector cells, such as NK cells and T cells, thereby influencing the generation and outcome of the antitumor immune response. Blood cancers can evade or dampen iNKT cell responses by downregulating expression of recognition receptors or by actively suppressing or diverting iNKT cell functions. This review will discuss literature on iNKT cell activity and associated dysregulation in blood cancers as well as highlight some of the strategies designed to harness and enhance iNKT cell functions against blood cancers.
Collapse
Affiliation(s)
- Pui Yeng Lam
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Michael D. Nissen
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Stephen R. Mattarollo
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
37
|
Dashtsoodol N, Shigeura T, Tashiro T, Aihara M, Chikanishi T, Okada H, Hanada K, Sano H, Kurogi A, Taniguchi M. Natural Killer T Cell-Targeted Immunotherapy Mediating Long-term Memory Responses and Strong Antitumor Activity. Front Immunol 2017; 8:1206. [PMID: 28993781 PMCID: PMC5622408 DOI: 10.3389/fimmu.2017.01206] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/12/2017] [Indexed: 12/19/2022] Open
Abstract
Current tumor therapies, including immunotherapies, focus on passive eradication or at least reduction of the tumor mass. However, cancer patients quite often suffer from tumor relapse or metastasis after such treatments. To overcome these problems, we have developed a natural killer T (NKT) cell-targeted immunotherapy focusing on active engagement of the patient’s immune system, but not directly targeting the tumor cells themselves. NKT cells express an invariant antigen receptor α chain encoded by Trav11 (Vα14)-Traj18 (Jα18) gene segments in mice and TRAV10 (Vα24)-TRAJ18 (Jα18) in humans and recognize glycolipid ligand in conjunction with a monomorphic CD1d molecule. The NKT cells play a pivotal role in the orchestration of antitumor immune responses by mediating adjuvant effects that activate various antitumor effector cells of both innate and adaptive immune systems and also aid in establishing a long-term memory response. Here, we established NKT cell-targeted therapy using a newly discovered NKT cell glycolipid ligand, RK, which has a stronger capacity to stimulate both human and mouse NKT cells compared to previous NKT cell ligand. Moreover, RK mediates strong adjuvant effects in activating various effector cell types and establishes long-term memory responses, resulting in the continuous attack on the tumor that confers long-lasting and potent antitumor effects. Since the NKT cell ligand presented by the monomorphic CD1d can be used for all humans irrespective of HLA types, and also because NKT cell-targeted therapy does not directly target tumor cells, this therapy can potentially be applied to all cancer patients and any tumor types.
Collapse
Affiliation(s)
- Nyambayar Dashtsoodol
- Laboratory for Immune Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Core Research Laboratory, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Tomokuni Shigeura
- Laboratory for Immune Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Takuya Tashiro
- Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata, Japan
| | - Minako Aihara
- Laboratory for Immune Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Toshihiro Chikanishi
- Laboratory for Immune Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hiromi Okada
- Laboratory for Immune Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Keigo Hanada
- Laboratory for Immune Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hirokazu Sano
- Laboratory for Immune Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Akihiko Kurogi
- Laboratory for Immune Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masaru Taniguchi
- Laboratory for Immune Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
38
|
Co-delivery of tumor-derived exosomes with alpha-galactosylceramide on dendritic cell-based immunotherapy for glioblastoma. Cancer Lett 2017; 411:182-190. [PMID: 28947140 DOI: 10.1016/j.canlet.2017.09.022] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 09/12/2017] [Accepted: 09/16/2017] [Indexed: 01/21/2023]
Abstract
Dendritic cell (DC) vaccine-based immunotherapy for glioblastoma multiforme (GBM) has shown apparent benefit in animal experiments and early-phase clinical trials, but the survival benefit is variable. In this work, we analyzed the mechanism of the potent antitumor immune response induced in vivo by tumor-associated antigen (TAA)-specific DCs with an invariant natural killer T (iNKT) cell adjuvant in orthotopic glioblastoma-bearing rats vaccinated with tumor-derived exosomes and α-galactosylceramide (α-GalCer) -pulsed DCs. Compared with traditional tumor lysate, exosomes were utilized as a more potent antigen to load DCs. iNKT cells, as an effective cellular adjuvant activated by α-GalCer, strengthened TAA presentation through their interaction with DCs. Co-delivery of tumor-derived exosomes with α-GalCer on a DC-based vaccine showed powerful effects in glioblastoma immunotherapy. This vaccine induced strong activation and proliferation of tumor-specific cytotoxic T lymphocytes, synergistically breaking the immune tolerance and improving the immunosuppressive environment.
Collapse
|
39
|
Goswami M, Hourigan CS. Novel Antigen Targets for Immunotherapy of Acute Myeloid Leukemia. Curr Drug Targets 2017; 18:296-303. [PMID: 25706110 DOI: 10.2174/1389450116666150223120005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) was the first malignancy for which immunotherapy, in the form of allogeneic hematopoietic stem cell transplantation (allo-HSCT), was integrated into the standard of care. Allo-HSCT however is an imperfect therapy associated with significant morbidity and mortality while offering only incomplete prevention of AML clinical relapse. These limitations have motivated the search for AML-related antigens that might be used as more specific and effective targets of immunotherapy. While historically such investigations have focused on protein targets expressed uniquely in AML or at significantly higher levels than in normal tissues, this article will review recent discoveries which have identified a novel selection of potential antigen targets for AML immunotherapy, such as non-protein targets including lipids and carbohydrates, neo-antigens created from genetic somatic mutations or altered splicing and post-translational modification of protein targets, together with innovative ways to target overexpressed protein targets presented by cell surface peptide-MHC complexes. These novel antigens represent promising candidates for further development as targets of AML immunotherapy.
Collapse
Affiliation(s)
- Meghali Goswami
- Myeloid Malignancies Section, National Heart, Lung and Blood Institute, Room 6C-104, 10 Center Drive, Bethesda, Maryland 20892-1583, United States
| | | |
Collapse
|
40
|
Abstract
Dendritic cells (DCs) play critical roles in activating innate immune cells and initiating adaptive immune responses. The functions of DCs were originally obscured by their overlap with other mononuclear phagocytes, but new mouse models have allowed for the selective ablation of subsets of DCs and have helped to identify their non-redundant roles in the immune system. These tools have elucidated the functions of DCs in host defense against pathogens, autoimmunity, and cancer. This review will describe the mouse models generated to interrogate the role of DCs and will discuss how their use has progressively clarified our understanding of the unique functions of DC subsets.
Collapse
Affiliation(s)
- Vivek Durai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
41
|
Fujii SI, Shimizu K. Exploiting Antitumor Immunotherapeutic Novel Strategies by Deciphering the Cross Talk between Invariant NKT Cells and Dendritic Cells. Front Immunol 2017; 8:886. [PMID: 28824620 PMCID: PMC5535079 DOI: 10.3389/fimmu.2017.00886] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 07/11/2017] [Indexed: 01/04/2023] Open
Abstract
Immune checkpoint blockade therapy has prevailed for several types of cancer; however, its effectiveness as a single therapy is still limited. In principle, dendritic cells (DCs) should be able to control the post-therapy immune response, in particular since they can link the two major arms of the immune system: innate and adaptive immunity. Therefore, DCs would be a logical and ideal target for the development of immunotherapies. Since DCs are not activated in the steady state, an adjuvant to convert their function from tolerogenic to immunogenic would be desirable. Upon ligand activation, invariant natural killer T (iNKT) cells simultaneously activate NK cells and also energize the DCs, resulting in their full maturation. To utilize such iNKT-licensed "fully" matured DCs as adjuvants, mechanisms of both intercellular communication between DC subsets and iNKT cells and intracellular molecular signaling in DCs have to be clarified and optimized. To generate both innate and adaptive immunity against cancer, a variety of strategies with the potential to target iNKT-licensed DCs in situ have been studied. The benchmark of success in these studies, each with distinct approaches, will be the development of functional NK cells and cytotoxic T cells (CTLs) as well as generation of long-term, memory CTL. In this review, we provide a framework for NKT-mediated immunotherapy through selective DC targeting in situ, describe progress in the design of licensed therapies for iNKT cell targeting of DCs, and highlight the challenge to provide maximal benefit to patients.
Collapse
Affiliation(s)
- Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| |
Collapse
|
42
|
Ghinnagow R, Cruz LJ, Macho-Fernandez E, Faveeuw C, Trottein F. Enhancement of Adjuvant Functions of Natural Killer T Cells Using Nanovector Delivery Systems: Application in Anticancer Immune Therapy. Front Immunol 2017; 8:879. [PMID: 28798749 PMCID: PMC5529346 DOI: 10.3389/fimmu.2017.00879] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/10/2017] [Indexed: 11/13/2022] Open
Abstract
Type I natural killer T (NKT) cells have gained considerable interest in anticancer immune therapy over the last decade. This “innate-like” T lymphocyte subset has the unique ability to recognize foreign and self-derived glycolipid antigens in association with the CD1d molecule expressed by antigen-presenting cells. An important property of these cells is to bridge innate and acquired immune responses. The adjuvant function of NKT cells might be exploited in the clinics. In this review, we discuss the approaches currently being used to target NKT cells for cancer therapy. In particular, we highlight ongoing strategies utilizing NKT cell-based nanovaccines to optimize immune therapy.
Collapse
Affiliation(s)
- Reem Ghinnagow
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - Luis Javier Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Elodie Macho-Fernandez
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - Christelle Faveeuw
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - François Trottein
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| |
Collapse
|
43
|
Dendritic cells combined with tumor cells and α-galactosylceramide induce a potent, therapeutic and NK-cell dependent antitumor immunity in B cell lymphoma. J Transl Med 2017; 15:115. [PMID: 28549432 PMCID: PMC5446707 DOI: 10.1186/s12967-017-1219-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/18/2017] [Indexed: 02/06/2023] Open
Abstract
Background Invariant natural killer T (iNKT) cells are a small population of lymphocytes with unique specificity for glycolipid antigens presented by non-polymorphic CD1d receptor on dendritic cells (DCs). iNKT cells play a central role in tumor immunology since they are implicated in the coordination of innate and adaptive immune responses. These cells can be activated with the prototypic lipid α-galactosylceramide (α-GalCer), stimulating interferon gamma (IFN-γ) production and cytokine secretion, which contribute to the enhancement of T cell activation. Methods We evaluated the antitumor effect of a combination of dendritic cells (DCs) and tumor cells with the iNKT cell agonist α-GalCer in a therapeutic model of B cell lymphoma. iNKT, NK and T cell phenotype was determined by flow cytometry. Serum cytokines were analyzed by Luminex technology. Significant differences between survival curves were assessed by the log-rank test. For all other data, Mann–Whitney test was used to analyze the differences between groups. Results This vaccine induced a potent (100% survival), long-lasting and tumor-specific antitumor immune response, that was associated with an increase of both Th1 cytokines and IFN-γ secreting iNKT cells (4.59 ± 0.41% vs. 0.92 ± 0.12% in control group; p = 0.01) and T cells (CD4 IFN-γ+: 3.75 ± 0.59% vs. 0.66 ± 0.18% p = 0.02; CD8 IFN-γ+: 10.61 ± 0.84% vs. 0.47 ± 0.03% p = 0.002). Importantly, natural killer (NK) cells played a critical role in the antitumor effect observed after vaccination. Conclusions This study provides clinically relevant data for the development of iNKT-cell based immunotherapy treatments for patients with B cell malignancies.
Collapse
|
44
|
Abstract
Conventional vaccine adjuvants enhance peptide-specific T-cell and B-cell responses by modifying peptide stability or uptake or by binding to pattern-recognition receptors on antigen-presenting cells (APCs). This article discusses the application of a distinct mechanism of adjuvant activity: the activation of type I, or invariant, natural killer T (iNKT) cells to drive cellular and humoral immune responses. Using a semi-invariant T-cell receptor (TCR), iNKT cells recognize glycolipid antigens presented on cluster of differentiation (CD)-1d molecules. When their ligands are presented in concert with peptides, iNKT cells can provide T-cell help, 'licensing' APCs to augment peptide-specific T-cell and antibody responses. We discuss the potential benefits and limitations of exploiting iNKT cells as 'universal helpers' to enhance vaccine responses for the treatment and prevention of cancer and infectious diseases.
Collapse
Affiliation(s)
- Mary Speir
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand.
- School of Biological Sciences, Victoria University Wellington, PO Box 600, Wellington, 6140, New Zealand.
- Maurice Wilkins Centre, Private Bag 92019, Auckland, New Zealand.
| | - Robert Weinkove
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand.
- Wellington Blood and Cancer Centre, Wellington Hospital, Private Bag 7902, Wellington, 6242, New Zealand.
- Department of Pathology and Molecular Medicine, University of Otago Wellington, Wellington, 6021, New Zealand.
| |
Collapse
|
45
|
Immunotherapeutic strategies targeting natural killer T cell responses in cancer. Immunogenetics 2016; 68:623-38. [PMID: 27393665 DOI: 10.1007/s00251-016-0928-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/22/2016] [Indexed: 12/21/2022]
Abstract
Natural killer T (NKT) cells are a unique subset of lymphocytes that bridge the innate and adaptive immune system. NKT cells possess a classic αβ T cell receptor (TCR) that is able to recognize self and foreign glycolipid antigens presented by the nonclassical class I major histocompatibility complex (MHC) molecule, CD1d. Type I NKT cells (referred to as invariant NKT cells) express a semi-invariant Vα14Jα18 TCR in mice and Vα24Jα18 TCR in humans. Type II NKT cells are CD1d-restricted T cells that express a more diverse set of TCR α chains. The two types of NKT cells often exert opposing effects especially in tumor immunity, where type II cells generally suppress tumor immunity while type I NKT cells can enhance anti-tumor immune responses. In this review, we focus on the role of NKT cells in cancer. We discuss their effector and suppressive functions, as well as describe preclinical and clinical studies utilizing therapeutic strategies focused on harnessing their potent anti-tumor effector functions, and conclude with a discussion on potential next steps for the utilization of NKT cell-targeted therapies for the treatment of cancer.
Collapse
|
46
|
Shimizu K, Yamasaki S, Shinga J, Sato Y, Watanabe T, Ohara O, Kuzushima K, Yagita H, Komuro Y, Asakura M, Fujii SI. Systemic DC Activation Modulates the Tumor Microenvironment and Shapes the Long-Lived Tumor-Specific Memory Mediated by CD8+ T Cells. Cancer Res 2016; 76:3756-66. [DOI: 10.1158/0008-5472.can-15-3219] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/04/2016] [Indexed: 11/16/2022]
|
47
|
Abstract
CD1- and MHC-related molecule-1 (MR1)-restricted T lymphocytes recognize nonpeptidic antigens, such as lipids and small metabolites, and account for a major fraction of circulating and tissue-resident T cells. They represent a readily activated, long-lasting population of effector cells and contribute to the early phases of immune response, orchestrating the function of other cells. This review addresses the main aspects of their immunological functions, including antigen and T cell receptor repertoires, mechanisms of nonpeptidic antigen presentation, and the current evidence for their participation in human and experimental diseases.
Collapse
Affiliation(s)
- Lucia Mori
- Department of Biomedicine, Basel University Hospital and Basel University, CH-4031 Basel, Switzerland; , , .,Singapore Immunology Network, A*STAR, 138648 Singapore
| | - Marco Lepore
- Department of Biomedicine, Basel University Hospital and Basel University, CH-4031 Basel, Switzerland; , ,
| | - Gennaro De Libero
- Department of Biomedicine, Basel University Hospital and Basel University, CH-4031 Basel, Switzerland; , , .,Singapore Immunology Network, A*STAR, 138648 Singapore
| |
Collapse
|
48
|
Godfrey DI, Uldrich AP, McCluskey J, Rossjohn J, Moody DB. The burgeoning family of unconventional T cells. Nat Immunol 2016; 16:1114-23. [PMID: 26482978 DOI: 10.1038/ni.3298] [Citation(s) in RCA: 582] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/15/2015] [Indexed: 02/07/2023]
Abstract
While most studies of T lymphocytes have focused on T cells reactive to complexes of peptide and major histocompatibility complex (MHC) proteins, many other types of T cells do not fit this paradigm. These include CD1-restricted T cells, MR1-restricted mucosal associated invariant T cells (MAIT cells), MHC class Ib-reactive T cells, and γδ T cells. Collectively, these T cells are considered 'unconventional', in part because they can recognize lipids, small-molecule metabolites and specially modified peptides. Unlike MHC-reactive T cells, these apparently disparate T cell types generally show simplified patterns of T cell antigen receptor (TCR) expression, rapid effector responses and 'public' antigen specificities. Here we review evidence showing that unconventional T cells are an abundant component of the human immune system and discuss the immunotherapeutic potential of these cells and their antigenic targets.
Collapse
Affiliation(s)
- Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, Australia
| | - Adam P Uldrich
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, Australia
| | - James McCluskey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia.,Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Australia
| | - D Branch Moody
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
49
|
Jiang Z, Zhang H, Wang Y, Yu B, Wang C, Liu C, Lu J, Chen F, Wang M, Yu X, Lin J, Pan X, Wang P, Zhu H. Altered Hepa1-6 cells by dimethyl sulfoxide (DMSO)-treatment induce anti-tumor immunity in vivo. Oncotarget 2016; 7:9340-52. [PMID: 26824185 PMCID: PMC4891044 DOI: 10.18632/oncotarget.7009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 01/01/2016] [Indexed: 12/30/2022] Open
Abstract
Cancer immunotherapy is the use of the immune system to treat cancer. Our current research proposed an optional strategy of activating immune system involving in cancer immunotherapy. When being treated with 2% DMSO in culture medium, Hepa1-6 cells showed depressed proliferation with no significant apoptosis or decreased viability. D-hep cells, Hepa1-6 cells treated with DMSO for 7 days, could restore to the higher proliferation rate in DMSO-free medium, but alteration of gene expression profile was irreversible. Interestingly, tumors from D-hep cells, not Hepa1-6 cells, regressed in wild-type C57BL/6 mice whereas D-hep cells exhibited similar tumorigenesis as Hep1–6 cells in immunodeficient mice. As expected, additional Hepa1-6 cells failed to form tumors in the D-hep-C57 mice in which D-hep cells were eliminated. Further research confirmed that D-hep-C57 mice established anti-tumor immunity against Hepa1-6 cells. Our research proposed viable tumor cells with altered biological features by DMSO-treatment could induce anti-tumor immunity in vivo.
Collapse
Affiliation(s)
- Zhengyu Jiang
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China.,Department of Anesthesiology, Second Military Medical University, Shanghai, P.R. China
| | - Hongxia Zhang
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Ye Wang
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Bin Yu
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Chen Wang
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Changcheng Liu
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Juan Lu
- Training Department, Second Military Medical University, Shanghai, P.R. China
| | - Fei Chen
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Minjun Wang
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Xinlu Yu
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Jiahao Lin
- School of Clinic Medicine, Second Military Medical University, Shanghai, P.R. China
| | - Xinghua Pan
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Pin Wang
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, P.R. China
| | - Haiying Zhu
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| |
Collapse
|
50
|
Seledtsov VI, Goncharov AG, Seledtsova GV. Clinically feasible approaches to potentiating cancer cell-based immunotherapies. Hum Vaccin Immunother 2016; 11:851-69. [PMID: 25933181 DOI: 10.1080/21645515.2015.1009814] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The immune system exerts both tumor-destructive and tumor-protective functions. Mature dendritic cells (DCs), classically activated macrophages (M1), granulocytes, B lymphocytes, aβ and ɣδ T lymphocytes, natural killer T (NKT) cells, and natural killer (NK) cells may be implicated in antitumor immunoprotection. Conversely, tolerogenic DCs, alternatively activated macrophages (M2), myeloid-derived suppressor cells (MDSCs), and regulatory T (Tregs) and B cells (Bregs) are capable of suppressing antitumor immune responses. Anti-cancer vaccination is a useful strategy to elicit antitumor immune responses, while overcoming immunosuppressive mechanisms. Whole tumor cells or lysates derived thereof hold more promise as cancer vaccines than individual tumor-associated antigens (TAAs), because vaccinal cells can elicit immune responses to multiple TAAs. Cancer cell-based vaccines can be autologous, allogeneic or xenogeneic. Clinical use of xenogeneic vaccines is advantageous in that they can be most effective in breaking the preexisting immune tolerance to TAAs. To potentiate immunotherapy, vaccinations can be combined with other modalities that target different immune pathways. These modalities include 1) genetic or chemical modification of cell-based vaccines; 2) cross-priming TAAs to T cells by engaging dendritic cells; 3) T-cell adoptive therapy; 4) stimulation of cytotoxic inflammation by non-specific immunomodulators, toll-like receptor (TLR) agonists, cytokines, chemokines or hormones; 5) reduction of immunosuppression and/or stimulation of antitumor effector cells using antibodies, small molecules; and 6) various cytoreductive modalities. The authors envisage that combined immunotherapeutic strategies will allow for substantial improvements in clinical outcomes in the near future.
Collapse
Key Words
- ADCC, antibody-dependent cell cytotoxicity
- APC, antigen-presenting cell
- Ab, antibodies
- BCG, Bacillus Calmette-Guérin
- Breg, regulatory B cell
- CAR, chimeric antigen receptor
- COX, cyclooxygenase
- CTA, cancer/testis antigen
- CTL, cytotoxic T lymphocyte
- CTLA-4, cytotoxic T lymphocyte antigen-4
- DC, dendritic cell
- DTH, delayed-type hypersensitivity
- GITR, glucocorticoid-induced tumor necrosis factor receptor
- GM-CSF, granulocyte-macrophage colony stimulating factor
- HIFU, high-intensity focused ultrasound
- IDO, indoleamine-2, 3-dioxygenase
- IFN, interferon
- IL, interleukin
- LAK, lymphokine-activated killer
- M, macrophage
- M1, classically activated macrophage
- M2, alternatively activated macrophage, MDSC, myeloid-derived suppressor cell
- MHC, major histocompatibility complex
- NK, natural killer (cell)
- PD-1, programmed death-1
- PGE2, prostaglandin E2
- RFA, radiofrequency ablation
- RNS, reactive nitrogen species
- ROS
- TAA, tumor-associated antigen
- TGF, transforming growth factor
- TLR, toll-like receptor
- TNF, tumor necrosis factor
- Th, T-helper cell
- Treg, regulatory T cell
- VEGF, vascular endothelial growth factor
- antitumor immunoprotection
- cancer cell-based vaccines
- combined immunotherapy
- immunosuppression
- reactive oxygen species
Collapse
Affiliation(s)
- V I Seledtsov
- a lmmanuel Kant Baltic Federal University ; Kaliningrad , Russia
| | | | | |
Collapse
|