1
|
Cirella A, Olivera I, Luri-Rey C, Bolaños E, Berraondo P, Melero I. Interleukin-18 in cancer immunology and immunotherapy. Expert Opin Ther Targets 2023; 27:1035-1042. [PMID: 37993172 DOI: 10.1080/14728222.2023.2287574] [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: 09/05/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
INTRODUCTION Interleukin-18 (IL-18) is a myeloid leukocyte inflammatory mediator whose main known function is to elicit IFNγ secretion from T and NK cells. AREAS COVERED This function offers potential in cancer immunotherapy but as a single treatment, preclinical and clinical antitumor activities are modest. IL-18 bioactivity is chiefly downregulated by a decoy soluble receptor named IL18-binding protein (IL-18BP) that is induced by IFNγ as a negative feedback mechanism. Recent advances indicate promising efficacy of IL-18 at armoring CAR-T cells for the treatment of hematological malignancies. Preclinical research has also yielded IL-18 constructs that do not bind IL-18BP but have preserved activity on the receptor and exert markedly increased antitumor effects. Indeed, agents of this kind are undergoing clinical trials. The synergistic effects of IL-18 and IL-12 in combination to induce IFNγ are extremely potent but are toxic if systemically delivered. In mouse models, IL-12 and decoy-resistant variants of IL-18 can be efficaciously used as local treatments for tumors by exploiting mRNA intratumoral co-delivery. Moreover, antitumor T cells can be transiently engineered with mRNAs encoding this combination of cytokines to attain efficacious synergistic effects also upon intratumoral delivery. EXPERT OPINION IL-18 certainly holds promise for immunotherapy in combination with other agents and for local approaches.
Collapse
Affiliation(s)
- Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Irene Olivera
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Elixabet Bolaños
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
- Department of Immunology and immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
- Department of oncology, Clínica Universidad de Navarra, Madrid, Spain
- Centro del Cancer de la Universidad de Navarra (CCUN), Pamplona, Spain
- Nuffield Department of Medicine (NDM), University of Oxford, Oxford, UK
| |
Collapse
|
2
|
Guan P, Schaub R, Nichols KE, Das R. Combination of NKT14m and Low Dose IL-12 Promotes Invariant Natural Killer T Cell IFN-γ Production and Tumor Control. Int J Mol Sci 2020; 21:ijms21145085. [PMID: 32708464 PMCID: PMC7404385 DOI: 10.3390/ijms21145085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/08/2020] [Accepted: 07/16/2020] [Indexed: 12/26/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are innate-like T lymphocytes characterized by the expression of an invariant T cell receptor (iTCR) that recognizes glycolipid antigens presented by the MHC I-like CD1d molecule. Following antigenic stimulation, iNKT cells rapidly produce large amounts of cytokines that can trans-activate dendritic cells (DC) and promote the anti-tumor functions of cytotoxic lymphocytes, such as natural killer (NK) and CD8 T cells. Additionally, iNKT cells can mediate robust and direct cytotoxicity against CD1d+ tumor targets. However, many tumors down-regulate CD1d and evade iNKT cell attack. To circumvent this critical barrier to iNKT cell anti-tumor activity, a novel monoclonal antibody (mAb), NKT14 has been recently developed. This agonistic antibody binds directly and specifically to the iTCR of murine iNKT cells. In the current study, we demonstrate that NKT14m mediates robust activation, cytokine production and degranulation of murine iNKT cells, in vitro. Consistently, NKT14m also promoted iNKT cell activation and immunomodulatory functions, in vivo. Finally, administration of NKT14m with low dose interleukin (IL)-12 further augmented iNKT cell IFN-γ production in vivo, and this combination conferred superior suppression of tumor cell growth compared to NKT14m or IL-12 alone. Together, these data demonstrate that a combination treatment consisting of low dose IL-12 and iTCR-specific mAb may be an attractive alternative to activate iNKT cell anti-tumor functions.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigens, CD1d/immunology
- Cell Line, Tumor
- Cytokines/metabolism
- Cytotoxicity, Immunologic/drug effects
- Drug Synergism
- Drug Therapy, Combination/methods
- Immunomodulation/drug effects
- Interferon-gamma/metabolism
- Interleukin-12/pharmacology
- Lymphoma/drug therapy
- Lymphoma/immunology
- Mice
- Mice, Inbred C57BL
- Natural Killer T-Cells/drug effects
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/metabolism
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Peng Guan
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Robert Schaub
- RGS Consulting, 118 Jeremy Hill Road Pelham, Pelham, NH 03076, USA;
| | - Kim E. Nichols
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Rupali Das
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Correspondence: ; Tel.: +1-517-884-5049; Fax: +1-517-355-5125
| |
Collapse
|
3
|
Abstract
Cytokine-based immunotherapy is executed by harnessing cytokines to activate the immune system to suppress tumors. Th1-type cytokines including IL-1, IL-2, IL-12 and granulocyte-macrophage colony-stimulating factor are potent stimulators of Th1 differentiation and Th1-based antitumor response. Many preclinical studies demonstrated the antitumor effects of Th1 cytokines but their clinical efficacy is limited. Multiple factors influence the efficacy of immunotherapy for tumors. For instance immunosuppressive cells in the tumor microenvironment can produce inhibitory cytokines which suppress antitumor immune response. Most studies on cytokine immunotherapy focused on how to boost Th1 response; many studies combined cytokine-based therapy with other treatments to reverse immunosuppression in tumor microenvironment. In addition, cytokines have pleiotropic functions and some cytokines show paradoxical activities under different settings. Better understanding the physiological and pathological functions of cytokines helps clinicians to design Th1-based cancer therapy in clinical practice.
Collapse
Affiliation(s)
- Hong-Mei Xu
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, United Kingdom.
| |
Collapse
|
4
|
Palma G, Barbieri A, Bimonte S, Palla M, Zappavigna S, Caraglia M, Ascierto PA, Ciliberto G, Arra C. Interleukin 18: friend or foe in cancer. Biochim Biophys Acta Rev Cancer 2013; 1836:296-303. [PMID: 24120852 DOI: 10.1016/j.bbcan.2013.09.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 09/24/2013] [Accepted: 09/28/2013] [Indexed: 01/17/2023]
Abstract
In the last few years, the field of tumor immunology has significantly expanded and its boundaries, never particularly clear, have become less distinct. Although the immune system plays an important role in controlling tumor growth, it has also become clear that tumor growth can be promoted by inflammatory immune responses. A good example that exemplifies the ambiguous role of the immune system in cancer progression is represented by interleukin 18 (IL-18) that was first identified as an interferon-γ-inducing factor (IGIF) involved in T helper type-1 (Th1) immune response. The expression and secretion of IL-18 have been observed in various cell types from immune cells to circulating cancer cells. In this review we highlighted the multiple roles played by IL-18 in immune regulation, cancer progression and angiogenesis and the clinical potential that may result from such understanding.
Collapse
Affiliation(s)
- Giuseppe Palma
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale per lo studio e la cura dei tumori "Fondazione Giovanni Pascale" - IRCCS, Italy; Istituto Endocrinologia e Oncologia Sperimentale - Consiglio Nazionale delle Ricerche, Via Pansini, 80131 Naples, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Nie Y, Zhang ZR, He B, Gu Z. Investigation of PEG-PLGA-PEG nanoparticles-based multipolyplexes for IL-18 gene delivery. J Biomater Appl 2011; 26:893-916. [PMID: 21273262 DOI: 10.1177/0885328210384889] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nanoparticles were formulated with biodegradable monomethoxy (poly ethylene glycol)-poly(lactide-co-glycolide)-monomethoxy (poly ethylene glycol) of three different proportional (PEG-PLGA-PEG, lactic acid: glycolic acid = 80/20, 70/30, 50/50) and the cytotoxicity of nanoparticle was characterized according to US Pharmacopoeia XXIII recommendations on various cell lines, including L929, Chang's hepatocytes, primary mouse myoblasts, osteoblasts, and renal vascular endothelial cells. mIL-18 gene was first condensed by polycationic peptide polylysine (PLL), and then encapsulated in the PEG-PLGA-PEG NPs as a novel multi-polyplex gene delivery system - Polymer-PLL-DNA. (PPDs) After lyopholization, the morphology, particle size, zeta potential, and the integrity of DNA in the NPs were investigated. The expression of mIL-18 gene on CT-26 cells in vitro were determined by western blot, while in vivo efficacy was evaluated by tumor inhibition rate, histological section, and survival curve in pulmonary metastasis of colon cancer in BALB/c mice model. Results showed that the cytotoxicity of blank nanoparticles was related to the degradation properties of the polymers with different compositions. The NPs with LA:GA = 70/30 (NPs-73) was optimal for intravenous injection due to its low cytotoxicity. Physicochemical properties of the PPDs were not changed during the lyopholization, while mIL-18 was successfully expressed in vitro. The anti-tumor efficacy in vivo of PPDs showed improvement especially combined with chemotherapy of cisplatin, and confirmed the promising application of the PPDs system, which compared with any single treatment.
Collapse
Affiliation(s)
- Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China
| | | | | | | |
Collapse
|
6
|
Xu Q, Zhang X, Yue J, Liu C, Cao C, Zhong H, Ma Q. Human TGFalpha-derived peptide TGFalphaL3 fused with superantigen for immunotherapy of EGFR-expressing tumours. BMC Biotechnol 2010; 10:91. [PMID: 21176167 PMCID: PMC3018390 DOI: 10.1186/1472-6750-10-91] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Accepted: 12/22/2010] [Indexed: 11/10/2022] Open
Abstract
Background Monoclonal antibodies have been employed as targeting molecules of superantigen for the preclinical treatment of a variety of tumours. However, other targeting molecules, such as tumour-related ligands or peptides, are less exploited. Here, we tested other targeting molecules by genetically fusing the third loop of transforming growth factor alpha (TGFalphaL3) to mutant staphylococcal enterotoxin A (SEAD227A). Results The resultant fusion proteins were expressed in E. coli and purified to homogeneity through a Ni-NTA affinity column. Fusion protein TGFalphaL3SEAD227A can promote splenocyte proliferation to a level comparable to recombinant SEA (rSEA) and bind to EGFR-expressing tumour cells in an EGFR-dependent way. Consistent with these observations, TGFalphaL3SEAD227A exerted an inhibitory effect on the growth of EGFR-expressing tumour cells both in vitro and in vivo. Notably, significant infiltrations of CD8+ and CD4+ T cells were detected in the tumour tissues of these C57BL/6 mice treated with TGFalphaL3SEAD227A, suggesting the involvement of T cells in this tumour-inhibitory process. Conclusions The data here showed that TGFαL3 is capable of targeting superantigen to tumours and exerting an inhibitory effect on tumour growth, which enables TGFαL3SEAD227A to be an attractive candidate for the immunotherapy of EGFR-expressing tumours.
Collapse
Affiliation(s)
- Quanbin Xu
- Beijing Institute of Biotechnology, Taiping Road 27, Beijing, PR China.
| | | | | | | | | | | | | |
Collapse
|
7
|
Oncolytic adenovirus expressing interleukin-18 induces significant antitumor effects against melanoma in mice through inhibition of angiogenesis. Cancer Gene Ther 2010; 17:28-36. [PMID: 19498459 DOI: 10.1038/cgt.2009.38] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
It has been shown that interleukin 18 (IL-18) exerts antitumor activity. In this study, we investigated whether oncolytic adenovirus-mediated gene transfer of IL-18 could induce strong antitumor activity. A tumor-selective replicating adenovirus expressing IL-18 (ZD55-IL-18) was constructed by insertion of an IL-18 expression cassette into the ZD55 vector, which is based on deletion of the adenoviral E1B 55-kDa gene. It has been shown that ZD55-IL-18 exerted a strong cytopathic effect and significant apoptosis in tumor cells. ZD55-IL-18 significantly decreased vascular endothelial growth factor and CD34 expression in the melanoma cells. Treatment of established tumors with ZD55-IL-18 showed much stronger antitumor activity than that induced by ZD55-EGFP (enhanced green fluorescent protein) or Ad-IL-18. These data indicated that oncolytic adenovirus expressing IL-18 could exert potential antitumor activity through inhibition of angiogenesis and offer a novel approach to melanoma therapy.
Collapse
|
8
|
Adenovirus-mediated LIGHT gene modification in murine B-cell lymphoma elicits a potent antitumor effect. Cell Mol Immunol 2010; 7:296-305. [PMID: 20418899 DOI: 10.1038/cmi.2010.15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Here, we investigated the antitumor effect of adenovirus-mediated gene transfer of LIGHT, the tumor-necrosis factor (TNF) superfamily member also known as TNFSF14, in the murine A20 B-cell lymphoma. LIGHT gene modification resulted in upregulated expression of Fas and the accessory molecule--intercellular adhesion molecule-1 (ICAM-1) on A20 cells and led to enhanced A20 cell apoptosis. LIGHT-modified A20 cells effectively stimulated the proliferation of T lymphocytes and interferon (IFN)-gamma production in vitro. Immunization of BALB/c mice with a LIGHT-modified A20 cell vaccine efficiently elicited protective immunity against challenge with the parental tumor cell line. Adenovirus-mediated gene transfer of LIGHT by intratumoral injection exerted a very potent antitumor effect against pre-existing A20 cell lymphoma in BALB/c mice. This adenovirus-mediated LIGHT therapy induced substantial splenic natural killer (NK) and cytotoxic T lymphocyte (CTL) activity, enhanced tumor infiltration by inflammatory cells and increased chemokine expression of CC chemokine ligand 21 (CCL21), IFN-inducible protein-10 (IP-10) and monokine induced by IFN-gamma (Mig) from tumor tissues. Thus, adenovirus-mediated LIGHT therapy might have potential utility for the prevention and treatment of B-cell lymphoma.
Collapse
|
9
|
Pei DS, Zheng JN. Oncolytic adenoviruses expressing interleukin: a novel antitumour approach. Expert Opin Biol Ther 2010; 10:917-26. [DOI: 10.1517/14712598.2010.481668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
10
|
Intratumoral delivery of IL-18 naked DNA induces T-cell activation and Th1 response in a mouse hepatic cancer model. BMC Cancer 2007; 7:87. [PMID: 17519043 PMCID: PMC1903361 DOI: 10.1186/1471-2407-7-87] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Accepted: 05/23/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The novel cytokine, interleukin (IL)-18, is a strong interferon-gamma inducer and costimulatory factor in Th1 cell activation. IL-18 triggers IFN-gamma production and enhances cytolytic activity in both T and NK cells. However, the exact mechanism of antitumor action of IL-18 remains to be clarified. To determine the effects of IL-18 plasmid DNA on hepatic cancer in mice, CT26 murine colon adenocarcinoma cells were established in mouse liver. METHODS Plasmid vectors encoding IL-18 were transferred directly into the liver 7 days after tumor injection to restrict IL-18 expression within the tumor site. The IL-18 protein level was increased in the liver 4 days after plasmid injection, and a marked antitumoral effect was observed at day 7. Antitumor effects were evaluated by measuring tumor regression, immune cell population, and IFN-gamma production. RESULTS The IL-18 plasmid controlled the growth of hepatic tumors and proliferation of splenic immune cells. Moreover, treatment of CT26 tumors with the IL-18 plasmid significantly enhanced the population of the effector T and NK cells in the spleen and peripheral blood. In spleen, the population of CD4+CD62Low cells was augmented in response to IL-18 on day 7. These results are consistent with the increase in CD4+ T cells secreting IFN-gamma, but not CD8+ T cells. The marked reduction of tumor growth in tumor-bearing mice was associated with the maintenance of IFN-gamma production in spleen in response to IL-18. These antitumoral effects were maintained until 14 days after plasmid injection. CONCLUSION Our results suggest that direct plasmid DNA transfer of IL-18 with no accompanying reagents to augment transfection efficiency may be useful in tumor immunotherapy.
Collapse
|
11
|
Zhang J, Wang Q, Zhao D, Cao X. Induction of potent anti-tumor immunity by direct injection of Ad-LIGHT at the site of tumor inoculation. Cytotherapy 2007; 9:386-96. [PMID: 17573614 DOI: 10.1080/14653240701326749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND The aim of this study was to observe the therapeutic effects of adenovirus-mediated LIGHT gene transfer in murine B16 melanoma in vivo. METHODS C57BL/6 mice were inoculated subcutaneously with B16 cells to establish the murine melanoma model. The tumor-bearing mice were injected at the site of tumor inoculation with recombinant adenoviral vectors expressing the murine LIGHT gene. The tumor growth and survival period of tumor-bearing mice were observed. The splenic NK and CTL activity were measured in vitro by lactate dehydrogenase (LDH) release assay. The amounts of cytokines were determined with ELISA kits. RESULTS The LIGHT gene could be efficiently transduced into tumor tissue after injection of Ad-LIGHT. Treatment with Ad-LIGHT significantly inhibited the tumor growth and prolonged the survival period of the tumor-bearing mice. The splenic NK and CTL activity of the mice was also enhanced after LIGHT gene transfer. The production of IL-2 and IFN-gamma from lymphocytes derived from mice treated with Ad-LIGHT was increased significantly compared with control groups. DISCUSSION Our results indicate that local expression of the LIGHT gene can induce potent anti-tumor immunity and may be a promising treatment strategy for melanoma.
Collapse
Affiliation(s)
- J Zhang
- Department of Medical Microbiology and Immunology, Medical School, Shaoxing University, Shaoxing, P. R. China.
| | | | | | | |
Collapse
|
12
|
Dai S, Zhou X, Wang B, Wang Q, Fu Y, Chen T, Wan T, Yu Y, Cao X. Enhanced induction of dendritic cell maturation and HLA-A*0201-restricted CEA-specific CD8(+) CTL response by exosomes derived from IL-18 gene-modified CEA-positive tumor cells. J Mol Med (Berl) 2006; 84:1067-76. [PMID: 17016692 PMCID: PMC7079873 DOI: 10.1007/s00109-006-0102-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2006] [Accepted: 07/05/2006] [Indexed: 01/20/2023]
Abstract
Dendritic cells (DC)-derived or tumor-derived exosomes are a population of nanometer sized membrane vesicles that can induce specific anti-tumor immunity. However, the immunogenic potential and efficiency of exosomes-based tumor vaccine are not satisfactory enough to achieve a curative effect in clinical trials. In this article we investigated whether IL-18 genetic modification of tumor cells can increase the efficacy of exosomes derived from IL-18 gene-modified tumor cells. We transfected carcinoembryonic antigen (CEA)-expressing tumor cells with a recombinant adenovirus encoding human IL-18 (AdhIL-18) and prepared the exosomes, Exo/IL-18, from IL-18 gene-modified tumor cells. We found that Exo/IL-18 naturally contain CEA and bioactive IL-18. Moreover, Exo-IL-18 are potent in chemoattracting DC and T cells, enhancing the proliferation and Th1 cytokine release of PBMC, and promoting the phenotypic and functional maturation of DC. Furthermore, Exo/IL-18-pulsed DC are quite potent to induce HLA-A*0201-restricted, CEA-specific CD8(+) CTL from the PBMC of HLA-A*0201 CEA(+) cancer patients in vitro. In almost all of these experiments, Exo/IL-18 show more potent functions than the conventionally prepared exosomes derived from parent tumor cells without IL-18 gene modification. Our findings suggest that Exo/IL-18 has more potent capability to induce specific anti-tumor immunity, and our strategy of IL-18 modification of exosomes is a feasible approach to develop exosomes-based tumor vaccines.
Collapse
Affiliation(s)
- Shengming Dai
- Institute of Immunology, Zhejiang University, Hangzhou, 310031 People’s Republic of China
- Institute of Immunology and State Key Laboratory of Medical Immunology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433 People’s Republic of China
| | - Xiangyang Zhou
- Institute of Immunology and State Key Laboratory of Medical Immunology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433 People’s Republic of China
| | - Baomei Wang
- Institute of Immunology and State Key Laboratory of Medical Immunology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433 People’s Republic of China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University, Hangzhou, 310031 People’s Republic of China
| | - Yangxin Fu
- Institute of Immunology, Zhejiang University, Hangzhou, 310031 People’s Republic of China
| | - Taoyong Chen
- Institute of Immunology and State Key Laboratory of Medical Immunology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433 People’s Republic of China
| | - Tao Wan
- Institute of Immunology and State Key Laboratory of Medical Immunology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433 People’s Republic of China
| | - Yizhi Yu
- Institute of Immunology and State Key Laboratory of Medical Immunology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433 People’s Republic of China
| | - Xuetao Cao
- Institute of Immunology, Zhejiang University, Hangzhou, 310031 People’s Republic of China
- Institute of Immunology and State Key Laboratory of Medical Immunology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433 People’s Republic of China
| |
Collapse
|
13
|
Marshall DJ, Rudnick KA, McCarthy SG, Mateo LRS, Harris MC, McCauley C, Snyder LA. Interleukin-18 enhances Th1 immunity and tumor protection of a DNA vaccine. Vaccine 2006; 24:244-53. [PMID: 16135392 DOI: 10.1016/j.vaccine.2005.07.087] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Accepted: 07/29/2005] [Indexed: 11/19/2022]
Abstract
DNA vaccines show efficacy in many preclinical models, but these results have not yet translated to consistent clinical efficacy. Co-administration of molecularly encoded adjuvants is one approach that may enable DNA vaccines to achieve enhanced immune response induction in humans. Interleukin-18 (IL-18) is a Th1-type cytokine that has been shown to augment the activity of DNA vaccines in some preclinical models. A prostate-specific antigen (PSA) DNA vaccine was tested in a mouse tumor model system to explore the impact of co-administration of a pIL-18 plasmid. Low doses of the pPSA vaccine were not capable of inducing tumor protection, but when pIL-18 was co-administered, complete tumor protection was observed in all mice. Tumor protection was mediated by both CD4(+) and CD8(+) T cells. Detailed analysis of the immune response in mice immunized with either pPSA or pPSA/pIL-18 demonstrated that pIL-18 skewed the PSA-specific immune response toward Th1. More importantly, stronger CD4(+) and CD8(+) T cell responses developed in the pPSA/pIL-18-immunized mice, with faster kinetics. These results suggest that IL-18 is a powerful adjuvant molecule that can enhance the development of antigen-specific immunity and vaccine efficacy.
Collapse
|
14
|
Huang C, Yu H, Wang Q, Yang G, Ma W, Xia D, Chen X, Yi P, Shen F, Zheng H, Cao X. A novel anticancer approach: SEA-anchored tumor cells expressing heat shock protein 70 onto the surface elicit strong anticancer efficacy. Immunol Lett 2005; 101:71-80. [PMID: 15908014 DOI: 10.1016/j.imlet.2005.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2005] [Accepted: 04/11/2005] [Indexed: 10/25/2022]
Abstract
Heat shock proteins (HSP) are attractive for their initiation of anticancer specific immunity via a distinct mechanism. To facilitate the induction process, we targeted HSP onto vaccine cell surface genetically. Then, SEA (a typical superantigen) was anchored on the cells by its fusion protein with transmembrane sequence, in order to produce immune-activated microsurrounding for further improvement of specific immunity. Thereby, the dual-modified vaccine, the surface-targeting-HSP70 and SEA-anchored vaccine, was developed successfully. Both in a therapeutic setting and in a pre-immune model, the mice vaccinated with the dual-modified vaccine displayed significant lymphocyte proliferation, higher NK and CTL activity, marked tumor suppression and prolonged survival when compared with those vaccinated with the vaccine modified alone with surface-targeting HSP70 or the SEA-anchored vaccine. Of all the vaccines, the dual-modified vaccine generated the best therapeutic efficacy on melanoma-bearing mice, the strongest protection against melanoma challenge. These results suggested that the dual-modified vaccine could induce more potent anticancer specific immunity while non-specific immunity was augmented.
Collapse
MESH Headings
- Animals
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/biosynthesis
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Cell Membrane/genetics
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Enterotoxins/metabolism
- Female
- HSP70 Heat-Shock Proteins/administration & dosage
- HSP70 Heat-Shock Proteins/biosynthesis
- HSP70 Heat-Shock Proteins/genetics
- HSP70 Heat-Shock Proteins/immunology
- Immunotherapy, Adoptive
- Male
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Transfection
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/biosynthesis
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
|
15
|
Sotiriadou NN, Perez SA, Gritzapis AD, Mahaira LG, Salagianni M, Baxevanis CN, Papamichail M. Beneficial effect of short-term exposure of human NK cells to IL15/IL12 and IL15/IL18 on cell apoptosis and function. Cell Immunol 2005; 234:67-75. [PMID: 16024007 DOI: 10.1016/j.cellimm.2005.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2005] [Revised: 05/15/2005] [Accepted: 05/19/2005] [Indexed: 10/25/2022]
Abstract
Monokines IL12, IL15, and IL18 have been shown to activate NK cell function, however with high apoptosis induced by their combination within 48 h. Here, we demonstrate for the first time that CD56+ cells incubated for only 18 h with the combination of IL15/IL12 or IL15/IL18, then washed, and further cultured in plain medium, exhibit low levels of apoptosis. These shortly activated CD56+ cells show high killer activity against NK- and LAK-sensitive tumor targets that persists over a culture period of 18 days after two additional 6 h cycles of exposure to the monokines applied every 8 days and also retain their ability for high cytokine production during each exposure. Moreover, these repetitive short-term exposures of CD56+ cells to the monokine combinations result in long-lived CD56+ cells with slower rates of FcgammaRIII receptor (CD16) decline, therefore exhibiting higher antibody depended cytotoxicity, as opposed to the continuous incubation with the monokine combinations. In conclusion, short-term exposure of CD56+ cells to IL15/IL12 or IL15/IL18 at 8-day intervals may hold a promise for improved clinical results in cellular adoptive cancer immunotherapy and for the in vivo injections of the monokines.
Collapse
|
16
|
Zhang B, Wu KF, Lin YM, Ma XT, Rao Q, Zheng GG, Cao ZY, Li G, Song YH. Gene transfer of pro-IL-18 and IL-1β converting enzyme cDNA induces potent antitumor effects in L1210 cells. Leukemia 2004; 18:817-25. [PMID: 14990975 DOI: 10.1038/sj.leu.2403320] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We report in a murine model of acute lymphoid leukemia L1210 the potent antitumor efficiency of a combinatorial delivery of pro-IL-18 gene modified L1210 (Lp18) and IL-1beta converting enzyme (ICE) gene modified L1210 (LpICE). Live leukemia cells Lp18 or Lp18 plus LpICE showed apparently reduced leukemogenicity with a survival rate of 40 or 50% at 50 days after intraperitoneal (i.p.) inoculation of a lethal dose of cells, respectively. Combination of Lp18 and LpICE was capable of inhibiting accumulation of bloody ascites, synergistically superior to Lp18 or LpICE alone. All surviving mice were rechallenged with parental L1210 cells at day 50, and all survived up to day 80, suggesting that gene-modified cells induced immune protection. Moreover, NK cytotoxicity and CTL activity were both enhanced in mice injected with Lp18, especially Lp18 plus LpICE. Levels of IFN-gamma were not altered significantly by inoculation of Lp18 or Lp18 plus LpICE. Our results demonstrate that IL-18 is a useful candidate gene in gene therapy of lymphoma or lymphoid leukemia, and ex vivo combinatorial delivery of Lp18 plus LpICE either as a single approach or as an adjunct to concomitant radiotherapy or chemotherapy, may be more efficient in a situation of minimal residual disease.
Collapse
Affiliation(s)
- B Zhang
- National Laboratory of Experimental Hematology, Institute of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Huang C, Yu H, Wang Q, Ma W, Xia D, Yi P, Zhang L, Cao X. Potent antitumor effect elicited by superantigen-linked tumor cells transduced with heat shock protein 70 gene. Cancer Sci 2004; 95:160-167. [PMID: 14965367 PMCID: PMC11159597 DOI: 10.1111/j.1349-7006.2004.tb03198.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2003] [Revised: 11/27/2003] [Accepted: 11/28/2003] [Indexed: 11/26/2022] Open
Abstract
Heat shock proteins (HSP) induce antitumor-specific immunity via a unique mechanism, but HSP alone fails to produce a satisfactory antitumor efficacy. We considered that the potent immune-activation of superantigen (SAg) might assist HSP to elicit a strong tumor-antigen-specific immunity. We initially prepared B16 melanoma cells linked to SAg SEA via a fusion protein with a transmembrane sequence (TM), and demonstrated that SEA thus anchored on the tumor cell surface could elicit strong antitumor immunity. We then prepared cells transduced with an inducible heat shock protein 70 (HSP70) gene, and bearing SEA-TM fusion protein on the cell surface, and used these cells as a dual-modified vaccine. In this study, either in a therapeutic setting or in a pre-immune model, the SEA-anchored vaccine or the HSP70 gene-modified vaccine induced marked tumor suppression, prolonged survival, augmented lymphocyte proliferation and higher NK and CTL activity in C57BL/6 mice compared with their controls (P < 0.01), though they were less effective than the dual-modified vaccine. Among these vaccines, the dual-modified vaccine showed the best therapeutic efficacy in B16 melanoma-bearing mice and gave the greatest protection against wild-type B16 melanoma challenge. The results indicated that the dual-modified vaccine could induce a potent tumor-antigen-specific immune response in addition to an increase of non-specific immunity. This study offers a novel approach to bridging specific and non-specific immunity for cancer therapy.
Collapse
Affiliation(s)
- Changxin Huang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Gutzmer R, Langer K, Mommert S, Wittmann M, Kapp A, Werfel T. Human dendritic cells express the IL-18R and are chemoattracted to IL-18. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:6363-71. [PMID: 14662834 DOI: 10.4049/jimmunol.171.12.6363] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IL-18 is secreted by a variety of cells such as epithelial cells, macrophages, and dendritic cells (DC), in particular, in areas of chronic inflammation. The effects of IL-18 are complex and not fully understood thus far. We sought to explore human DC as a new target for IL-18, since IL-18R expression has been described on myeloid cells such as macrophages and DC are likely to get in contact with IL-18 at sites of inflammatory reactions. We demonstrate the expression of the IL-18R on human DC in peripheral blood and epidermis, as well as monocyte-derived dendritic cells (MoDC). On MoDC, IL-18R expression is up-regulated by IFN-gamma. IL-18 strongly up-regulated CD54 on MoDC, whereas the effect on MHC class II, CD83, and CD86 was only moderate and the expression of CD40 and CD80 was not affected. MoDC primed with IL-18 did not increase their capacity to stimulate the proliferation or IFN-gamma production of autologous T cells. However, IL-18 had a direct migratory effect on MoDC as indicated by induction of filamentous actin polymerization and migration in Boyden chamber experiments. In epidermal DC, IL-18 was also able to induce filamentous actin polymerization. Therefore, IL-18 might represent a novel mechanism to recruit DC to areas of inflammation, in particular under Th1 cytokine conditions where IFN-gamma is increased such as psoriasis or inflammatory bowel diseases.
Collapse
Affiliation(s)
- Ralf Gutzmer
- Department of Dermatology and Allergology, Hannover Medical University, Hannover, Germany.
| | | | | | | | | | | |
Collapse
|
19
|
Baxevanis CN, Gritzapis AD, Papamichail M. In vivo antitumor activity of NKT cells activated by the combination of IL-12 and IL-18. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:2953-9. [PMID: 12960319 DOI: 10.4049/jimmunol.171.6.2953] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Interleukin-12 and IL-18 have been demonstrated to potentiate innate immunity in a variety of experimental tumor models, but the functional roles of NK and/or NKT cells and their mechanism of action in these models have not been fully addressed. Through adoptive transfer of NKT cells activated in vitro with a combination of IL-12 plus IL-18 (IL-12/IL-18 NKT) into syngeneic animals, we demonstrate in this study that IL-12/IL-18 NKT cells are essential and collaborate with the host's own NK cells in natural host immunity against the growth of ALC and MC57X syngeneic tumors. The relative roles of the adoptively transferred IL-12/IL-18 NKT cells and endogenous NK cells in host protection were first shown in normal C57BL/6 (B6) mice treated with anti-asialo GM1 Ab that selectively depletes NK cells; second, in B6.TCRJalpha281(-/-) mice specifically deficient for NKT cells; and third, in B6.scid mice that also lack NKT cells. Furthermore, by injecting normal B6 mice with anti-IL-2 and/or anti-IFN-gamma mAb, we could demonstrate that effective innate immunity against both types of syngeneic tumors was dependent on the production of IL-2 and IFN-gamma by the adoptively transferred NKT cells. In vitro studies confirmed both the secretion of IL-2 and IFN-gamma by the IL-12/IL-18-activated NKT cells and their collaborative role with NK cells for lysis of ALC and MC57X syngeneic tumor targets. This is the first description of an antitumor function of IL-12/IL-18 NKT cells adoptively transferred into syngeneic hosts that provides the basis for a new modality in the cellular immunotherapy of cancer.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens/biosynthesis
- Antigens, Surface
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- CD3 Complex/biosynthesis
- Cell Line, Tumor
- Cells, Cultured
- Cytotoxicity, Immunologic
- Fibrosarcoma/pathology
- Fibrosarcoma/prevention & control
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/pharmacology
- Interferon-gamma/biosynthesis
- Interferon-gamma/physiology
- Interleukin-12/administration & dosage
- Interleukin-12/pharmacology
- Interleukin-18/administration & dosage
- Interleukin-18/pharmacology
- Interleukin-2/biosynthesis
- Interleukin-2/physiology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/transplantation
- Lectins, C-Type
- Lymphocyte Activation/immunology
- Lymphoma, T-Cell/immunology
- Lymphoma, T-Cell/pathology
- Lymphoma, T-Cell/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, SCID
- NK Cell Lectin-Like Receptor Subfamily B
- Neoplasm Transplantation
- Protein Biosynthesis
- Proteins
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
Collapse
|
20
|
Xia D, Zheng S, Zhang W, He L, Wang Q, Pan J, Zhang L, Wang J, Cao X. Effective induction of therapeutic antitumor immunity by dendritic cells coexpressing interleukin-18 and tumor antigen. J Mol Med (Berl) 2003; 81:585-96. [PMID: 12937899 DOI: 10.1007/s00109-003-0472-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2003] [Accepted: 07/09/2003] [Indexed: 11/29/2022]
Abstract
Dendritic cell (DC) based cancer vaccine can induce potent antitumor immunity in murine models; however, objective clinical responses have been observed only in a minority of cancer patients. To improve the antitumor effect of DC vaccine, Th1-biasing cytokine interleukin (IL) 18 and melanoma-associated antigen gp100 were cotransfected into bone marrow-derived DC (IL-18/gp100-DC), which were used as vaccine to induce the protective and therapeutic immunity in a B16 melanoma model. Immunization with IL-18/gp100-DC resulted in tumor resistance in 87.5% of the mice challenged with B16 cells; however, 12.5% and 25% of mice immunized with gp100 gene-modified DC (gp100-DC) or IL-18 gene-modified DC (IL-18-DC) were tumor free, respectively. Most importantly, IL-18/gp100-DC immunization led to the generation of potent therapeutic immunity that significantly inhibited the tumor growth and improved the survival period of mice bearing established melanoma. Immune cell depletion experiments identified that CD4(+) T cells also played an important role in the priming phase of antitumor immunity and CD8(+) T lymphocytes were the primary effectors. gp100-specific CTL response were induced most markedly in the tumor-bearing mice immunized with IL-18/gp100-DC. Administration with such vaccine also significantly increased the production of Th1 cytokine (IL-2 and interferon-gamma) and induced infiltration of inflammatory cells inside and around the tumors. In addition, natural killer cell activity was also augmented. These results indicate that immunization with DC vaccine coexpressing Th1 cytokine IL-18 and tumor antigen gene may be an effective strategy for a successful therapeutic vaccination.
Collapse
Affiliation(s)
- Dajing Xia
- Institute of Immunology, Zhejiang University, 310031, Hangzhou, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Tanaka F, Hashimoto W, Robbins PD, Lotze MT, Tahara H. Therapeutic and specific antitumor immunity induced by co-administration of immature dendritic cells and adenoviral vector expressing biologically active IL-18. Gene Ther 2002; 9:1480-6. [PMID: 12378411 DOI: 10.1038/sj.gt.3301827] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2002] [Accepted: 05/31/2002] [Indexed: 11/08/2022]
Abstract
Interleukin-18 is a potent cytokine expressed early in the immune response following cleavage in activated composes. We have investigated the in vivo antitumor effects of intratumoral (i.t.) administration of an adenoviral vector expressing biologically active murine interleukin (IL)-18 (Ad.PTH.IL-18). Substantial antitumor effects were observed when established MCA205 fibrosarcoma was treated in syngeneic immunocompetent mice with intratumoral injection of Ad.PTH.IL-18 (P = 0.0025 versus control vector treatment), generating potent cytotoxic T lymphocytes (CTLs) in culture. In contrast, the antitumor effect was absent, and cytotoxic activity was significantly less (P = 0.021) in gld mice (Fas ligand deficient). To enhance the in vivo antitumor activity of the treatment using Ad.PTH.IL-18, we co-injected immature DC and Ad.PTH.IL-18 i.t. into established, day 7 MCA205 fibrosarcoma and MC38 adenocarcinoma. Co-injection of both Ad.PTH.IL-18 and DC was associated with complete abrogation of injected tumors. Furthermore, the antitumor effects were also observed on distant tumors inoculated i.d. in the contralateral flank of the animal. The induced cytolytic activity was tumor-specific and MHC class I-restricted. As we have previously demonstrated in vitro (Tanaka F et al, Cancer Res 2000; 60: 4838-4844) and consistent with these findings in vivo, NK, T and dendritic cells coactivately mediate the IL-18 enhanced antitumor effect. This study suggests that the coactivate strategy could be used in the clinical setting to treat patients with cancer. do
Collapse
Affiliation(s)
- F Tanaka
- Department of Surgery, University of Pittsburgh School of Medicine, PA, USA
| | | | | | | | | |
Collapse
|