1
|
Chen C, Zhou Z, Niu K, Du C, Liang A, Yang L. Efficacy and Safety of Nasal Immunisation with Somatostatin DNA Vaccine for Growth Promotion in Fattening Pigs. Animals (Basel) 2022; 12:3072. [PMID: 36428299 PMCID: PMC9686601 DOI: 10.3390/ani12223072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/04/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
This study aimed to evaluate the efficacy and safety of the SS DNA vaccine on growing pigs. Randomly, 147 pigs were divided into four groups, treatment 1 (T1, 3 × 109 CFU/mL, n = 39), T2 (3 × 108 CFU/mL, n = 35), T3 (3 × 107 CFU/mL, n = 35) and control group (phosphate-buffered saline, n = 38). All animals received two vaccinations separated by 45 days and the same diet and management. The results showed that all treatment groups (T1, T2 and T3) had significantly higher slaughter weight (d 185) than the Ctrl group (p < 0.05), and daily gain between 50 and 110 days of age was significantly higher in all treatment groups than in the Ctrl group (p < 0.05). Antibody-positive pigs have significantly higher daily weight gain than that in antibody-negative pigs (p < 0.05). The results of the meat quality analysis showed no significant changes between the P (antibody-positive pigs) and N (antibody-negative pigs) groups. Furthermore, the results showed that antibody titres at 110 and 185 days had a significant positive correlation with the daily weight gain (p < 0.05) and a significant negative correlation with the backfat thickness (p < 0.05). Evaluating the safety of vaccines by PCR amplification of target genes (GS/2SS), faecal, soil and water samples had no target genes detected by PCR amplification in these samples after 5 days, and no GS/2SS were detected in the blood and tissues for the experimental period. Moreover, no abnormalities were found in pathological sections of the P group compared with the N group. In conclusion, SS DNA vaccines can promote the growth of fattening pigs to a certain extent without altering the meat quality, and it has no effects on the safety of the surrounding environment.
Collapse
Affiliation(s)
- Chao Chen
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zichao Zhou
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Kaifeng Niu
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Chao Du
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Aixin Liang
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Province’s Engineering Research Center in Buffalo Breeding and Products, Wuhan 430070, China
| | - Liguo Yang
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Province’s Engineering Research Center in Buffalo Breeding and Products, Wuhan 430070, China
| |
Collapse
|
2
|
Haghighi D, Yazdani S, Farzanehpour M, Esmaeili Gouvarchinghaleh H. Combined extract of heated TC1, a heat-killed preparation of Lactobacillus casei and alpha-galactosyl ceramide in a mouse model of cervical cancer. Infect Agent Cancer 2022; 17:51. [PMID: 36127698 PMCID: PMC9487028 DOI: 10.1186/s13027-022-00464-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022] Open
Abstract
Background Nowadays, cancer is the leading cause of death among threats to humanity, necessitating prompt action and preparation. Cervical cancer is one of the most common cancers in women and is currently treated with surgery, radiation, chemotherapy, and immunotherapy, among other treatments. Current oncology approaches focused on the simultaneous development of safe and effective cancer multi-agent therapies. The present study aimed to evaluate the effects of a combined extracts of heated TC1, a heat-killed preparation of Lactobacilluscasei, and alpha-galactosyl ceramide (α-GalCer) in a mouse model of cervical cancer. Material and methods Cervical cancer in the mouse model was prepared by TC1 cells subcutaneous injection into the left flank of female C57BL/6 mouse aged 6–8 weeks (n = 80). After the appearance of the palpable tumor, the mice with cervical cancer were randomly devoted to 8 (ten-member) groups. The mice in some groups were treated with PBS, TC1 cell extract, L. casei extract, α-GalCer, and a combination of the mentioned treatments. Then, they were evaluated the splenocytes proliferation, lactate dehydrogenase production and nitric oxide. Moreover, IL-4, IFN-γ, and TGF-β cytokine levels of splenocytes supernatant the mice were measured. In all evaluations, a statistical difference of less than 0.05 (P ˂ 0.05) was considered as a significant level. Result The findings revealed that the combination therapy group (heated TC1 cell and L. casei extracts with α-GalCer) significantly increases the splenocytes proliferation (MTT) (0.358 ± 0.04 OD), LDH production (45.9 ± 2.3 U/L), NO rate (38.4 ± 2.8 µM), and IFN-γ cytokine level (46.6 ± 3.7 pg/ml) (P < 0.05). Also, observes a significantly reduces the production of IL-4 (11.6 ± 2.5 pg/ml) and TGF-β cytokines levels (7.8 ± 2.5 pg/ml) (P < 0.05) in comparison to the control group. Conclusion The study showed that combination therapy of L. casei and α-GalCer is an efficient treatment for cervical cancer in the mouse model.
Collapse
Affiliation(s)
- Dorsa Haghighi
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shaghayegh Yazdani
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.,Department of Microbiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahdieh Farzanehpour
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
3
|
He M, Yang T, Wang Y, Wang M, Chen X, Ding D, Zheng Y, Chen H. Immune Checkpoint Inhibitor-Based Strategies for Synergistic Cancer Therapy. Adv Healthc Mater 2021; 10:e2002104. [PMID: 33709564 DOI: 10.1002/adhm.202002104] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/26/2021] [Indexed: 12/16/2022]
Abstract
Immune checkpoint blockade therapy (ICBT) targeting checkpoints, such as, cytotoxic T-lymphocyte associated protein-4 (CTLA-4), programmed death-1 (PD-1), or programmed death-ligand 1 (PD-L1), can yield durable immune response in various types of cancers and has gained constantly increasing research interests in recent years. However, the efficacy of ICBT alone is limited by low response rate and immune-related side effects. Emerging preclinical and clinical studies reveal that chemotherapy, radiotherapy, phototherapy, or other immunotherapies can reprogramm immunologically "cold" tumor microenvironment into a "hot" one, thus synergizing with ICBT. In this review, the working principle and current development of various immune checkpoint inhibitors are summarized, while the interactive mechanism and recent progress of ICBT-based synergistic therapies with other immunotherapy, chemotherapy, phototherapy, and radiotherapy in fundamental and clinical studies in the past 5 years are depicted and highlighted. Moreover, the potential issues in current studies of ICBT-based synergistic therapies and future perspectives are also discussed.
Collapse
Affiliation(s)
- Mengying He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Tao Yang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Yuhan Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Mengyuan Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Xingye Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Dawei Ding
- Jiangsu Key Laboratory of Neuropsychiatric Diseases College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Yiran Zheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Huabing Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
- State Key Laboratory of Radiation Medicine and Protection Soochow University Suzhou 215123 China
| |
Collapse
|
4
|
Shermeh AS, Zahedifard F, Habibzadeh S, Taheri T, Rafati S, Seyed N. Evaluation of protection induced by in vitro maturated BMDCs presenting CD8 + T cell stimulating peptides after a heterologous vaccination regimen in BALB/c model against Leishmania major. Exp Parasitol 2021; 223:108082. [PMID: 33581108 DOI: 10.1016/j.exppara.2021.108082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 01/07/2021] [Accepted: 01/30/2021] [Indexed: 11/28/2022]
Abstract
Leishmaniasis is a complex vector-borne disease mediated by Leishmania parasite and a strong and long-lasting CD4+ Th1 and CD8+-T cell immunity is required to control the infection. Thus far multivalent subunit vaccines have met this requirement more promisingly. However several full protein sequences cannot be easily arranged in one construct. Instead, new emerging immune-informatics based epitope formulations surpass this restriction. Herein, we aimed to examine the protective potential of a dendritic cell based vaccine presenting epitopes to CD8+ and CD4+-T cells in combination with DNA vaccine encoding the same epitopes against murine cutaneous leishmaniasis. Immature DCs were loaded with epitopes (selected from parasite proteome) in vitro with or without CpG oligonucleotides and were used to immunize BALB/c mice. Peptide coding DNA was used to boost the system and immunological responses were evaluated after Leishmania (L.) major infectious challenge. The pre-challenge response to included epitopes was Th1 polarized which potentially lowered the infection at early time points post-challenge but not at later weeks. Collectively, DC prime-DNA boost was found to be a promising approach for Th1 polarization however the constituent epitopes undoubtedly make a significant contribution in the protection outcome of the vaccine.
Collapse
Affiliation(s)
- Atefeh Sadeghi Shermeh
- Immunotherapy and Leishmania Vaccine Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Farnaz Zahedifard
- Immunotherapy and Leishmania Vaccine Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Sima Habibzadeh
- Immunotherapy and Leishmania Vaccine Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Tahereh Taheri
- Immunotherapy and Leishmania Vaccine Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Sima Rafati
- Immunotherapy and Leishmania Vaccine Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Negar Seyed
- Immunotherapy and Leishmania Vaccine Research Department, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
5
|
Guo S, Xiao P, Li B, Wang W, Wang S, Lv T, Xu X, Chen C, Huang L, Li Z, Tang L, Peng L, Wang H. Co-immunizing with PD-L1 induces CD8 + DCs-mediated anti-tumor immunity in multiple myeloma. Int Immunopharmacol 2020; 84:106516. [PMID: 32334387 DOI: 10.1016/j.intimp.2020.106516] [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] [Received: 12/25/2019] [Revised: 03/24/2020] [Accepted: 04/13/2020] [Indexed: 12/24/2022]
Abstract
Tumor therapeutic vaccines have faced a challenge for effective protection against malignant tumors by inducing tumor-specific CD8+ T cell responses. Here, we designed a DNA vaccine containing a tumor-specific antigen of Dickkopf-1 (DKK-1) and an immune checkpoint of programmed death ligand 1 (PD-L1) delivered by PLGA/PEI nanoparticle-mediated delivery system for multiple myeloma therapy. Murine subcutaneous tumor model established with human DKK1 (hDKK-1)-SP2/0 cells were intramuscularly immunized with PLGA/PEI-pPD-L1/pDDK-1 vaccine and equal amount of control 3 times at 10 day-intervals. Compared with PLGA/PEI-pDKK1 immunization group, PLGA/PEI-pPD-L1/pDKK-1 co-immunization enhanced the induction and mature of CD11c+ DCs and CD8+CD11c+ DCs, and promoted antigen-specific Th1 responses and cytotoxic T lymphocyte (CTL) responses. The reduced tumor volume and weight as well as increased tumor inhibition rate were observed in PLGA/PEI-pPD-L1/pDKK-1 vaccine co-immunization group, indicated that the vaccine could effectively inhibit the tumor growth of multiple myeloma. The anti-tumor activity of PLGA/PEI-pPD-L1/pDKK-1 vaccine was abrogated by CD8 cell depletion accompanied with the reduced percentages of CD8+CD11c+ DCs and CD8+ T cells in the spleen and TILs. These results indicated that the anti-tumor efficacy of PLGA/PEI-pPD-L1/pDKK-1 vaccine was required for CD8+CD11c+ DCs-mediated CD8+ T cell immunity responses. This vaccine strategy may represent a potential and promising approach for hematological malignancy treatment.
Collapse
Affiliation(s)
- Shuli Guo
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Pengli Xiao
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Bo Li
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Wanli Wang
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Songyun Wang
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Tao Lv
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Xiaoyan Xu
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Cong Chen
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Lei Huang
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Zhi Li
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Li Tang
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Liang Peng
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Huirui Wang
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China.
| |
Collapse
|
6
|
Feng H, Nakajima N, Wu L, Yamashita M, Lopes TJS, Tsuji M, Hasegawa H, Watanabe T, Kawaoka Y. A Glycolipid Adjuvant, 7DW8-5, Enhances the Protective Immune Response to the Current Split Influenza Vaccine in Mice. Front Microbiol 2019; 10:2157. [PMID: 31620111 PMCID: PMC6759631 DOI: 10.3389/fmicb.2019.02157] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/02/2019] [Indexed: 12/13/2022] Open
Abstract
Vaccination is an effective strategy to control influenza disease. Adjuvants enhance the efficacy of vaccines, but few adjuvants are approved for human use, so novel, safe, and effective adjuvants are urgently needed. The glycolipid adjuvant 7DW8-5 has shown adjuvanticity to malaria vaccine; however, its adjuvant effect for seasonal influenza vaccine remains unknown. Here, we evaluated the adjuvanticity of 7DW8-5 to a quadrivalent split influenza vaccine in a mouse model. 7DW8-5 significantly enhanced virus-specific antibody production when administrated with influenza vaccine compared with that of vaccine alone; 10 μg of 7DW8-5 induced similar antibody levels to those induced by alum. Mouse body weight loss was reduced and, notably, the survival rate was increased in the vaccine plus 7DW8-5 group compared with that in the vaccine plus alum group. Our results indicate that the glycolipid 7DW8-5 is a promising adjuvant for influenza vaccine.
Collapse
Affiliation(s)
- Huapeng Feng
- Division of Virology, Department of Microbiology and Immunology, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Noriko Nakajima
- Department of Pathology, The National Institute of Infectious Diseases, Tokyo, Japan
| | - Li Wu
- Division of Virology, Department of Microbiology and Immunology, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Makoto Yamashita
- Division of Virology, Department of Microbiology and Immunology, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Tiago J S Lopes
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Affiliate of the Rockefeller University, New York, NY, United States
| | - Hideki Hasegawa
- Department of Pathology, The National Institute of Infectious Diseases, Tokyo, Japan
| | - Tokiko Watanabe
- Division of Virology, Department of Microbiology and Immunology, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, The Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Sheng Z, Chen H, Feng K, Gao N, Wang R, Wang P, Fan D, An J. Electroporation-Mediated Immunization of a Candidate DNA Vaccine Expressing Dengue Virus Serotype 4 prM-E Antigen Confers Long-Term Protection in Mice. Virol Sin 2019; 34:88-96. [PMID: 30790202 DOI: 10.1007/s12250-019-00090-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/17/2019] [Indexed: 12/13/2022] Open
Abstract
Dengue fever, caused by dengue viruses (DENVs), is a widespread mosquito-borne zoonotic disease; however, there is no available anti-dengue vaccine for worldwide use. In the current study, a DNA vaccine candidate (pV-D4ME) expressing prM-E protein of DENV serotype 4 (DENV-4) was constructed, and its immunogenicity and protection were evaluated in immunocompetent BALB/c mice. The pV-D4ME candidate vaccine induced effective humoral and cellular immunity of mice against DENV-4 in vivo when administered both at 50 μg and 5 μg through electroporation. Two weeks after receiving three immunizations, both doses of pV-D4ME DNA were shown to confer effective protection against lethal DENV-4 challenge. Notably, at 6 months after the three immunizations, 50 μg, but not 5 μg, of pV-D4ME could provide stable protection (100% survival rate) against DENV-4 lethal challenge without any obvious clinical signs. These results suggest that immunization with 50 μg pV-D4ME through electroporation could confer effective and long-term protection against DENV-4, offering a promising approach for development of a novel DNA vaccine against DENVs.
Collapse
Affiliation(s)
- Ziyang Sheng
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Hui Chen
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Kaihao Feng
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Na Gao
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Ran Wang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Peigang Wang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Dongying Fan
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China. .,Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, 100093, China.
| |
Collapse
|
8
|
Experimental Combined Immunotherapy of Tumours with Major Histocompatibility Complex Class I Downregulation. Int J Mol Sci 2018; 19:ijms19113693. [PMID: 30469401 PMCID: PMC6274939 DOI: 10.3390/ijms19113693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/14/2018] [Accepted: 11/17/2018] [Indexed: 12/20/2022] Open
Abstract
Combined immunotherapy constitutes a novel, advanced strategy in cancer treatment. In this study, we investigated immunotherapy in the mouse TC-1/A9 model of human papillomavirus type 16 (HPV16)-associated tumors characterized by major histocompatibility complex class I (MHC-I) downregulation. We found that the induction of a significant anti-tumor response required a combination of DNA vaccination with the administration of an adjuvant, either the synthetic oligodeoxynucleotide ODN1826, carrying immunostimulatory CpG motifs, or α-galactosylceramide (α-GalCer). The most profound anti-tumor effect was achieved when these adjuvants were applied in a mix with a one-week delay relative to DNA immunization. Combined immunotherapy induced tumor infiltration with various subsets of immune cells contributing to tumor regression, of which cluster of differentiation (CD) 8⁺ T cells were the predominant subpopulation. In contrast, the numbers of tumor-associated macrophages (TAMs) were not markedly increased after immunotherapy but in vivo and in vitro results showed that they could be repolarized to an anti-tumor M1 phenotype. A blockade of T cell immunoglobulin and mucin-domain containing-3 (Tim-3) immune checkpoint had a negligible effect on anti-tumor immunity and TAMs repolarization. Our results demonstrate a benefit of combined immunotherapy comprising the activation of both adaptive and innate immunity in the treatment of tumors with reduced MHC-I expression.
Collapse
|
9
|
Barros MR, de Melo CML, Barros MLCMGR, de Cássia Pereira de Lima R, de Freitas AC, Venuti A. Activities of stromal and immune cells in HPV-related cancers. J Exp Clin Cancer Res 2018; 37:137. [PMID: 29976244 PMCID: PMC6034319 DOI: 10.1186/s13046-018-0802-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023] Open
Abstract
The immune system is composed of immune as well as non-immune cells. As this system is a well-established component of human papillomavirus- (HPV)-related carcinogenesis, high risk human papillomavirus (hrHPV) prevents its routes and mechanisms in order to cause the persistence of infection. Among these mechanisms are those originated from stromal cells, which include the cancer-associated fibroblasts (CAFs), the myeloid-derived suppressor cells (MDSCs) and the host infected cells themselves, i.e. the keratinocytes. These types of cells play central role since they modulate immune cells activities to create a prosperous milieu for cancer development, and the knowledge how such interactions occur are essential for prognostic assessment and development of preventive and therapeutic approaches. Nevertheless, the precise mechanisms are not completely understood, and this lack of knowledge precluded the development of entirely efficient immunotherapeutic strategies for HPV-associated tumors. As a result, an intense work for attaining how host immune response works, and developing of effective therapies has been applied in the last decade. Based on this, this review aims to discuss the major mechanisms of immune and non-immune cells modulated by hrHPV and the potential and existing immunotherapies involving such mechanisms in HPV-related cancers. It is noticed that the combination of immunotherapies has been demonstrated to be essential for obtaining better results, especially because the possibility of increasing the modulating capacity of the HPV-tumor microenvironment has been shown to be central in strengthening the host immune system.
Collapse
Affiliation(s)
- Marconi Rego Barros
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Center of Biological Sciences, Federal University of Pernambuco, Cidade Universitária, Av. Prof Moraes Rego, 1235, Recife, PE CEP-50670-901 Brazil
| | - Cristiane Moutinho Lagos de Melo
- Laboratory of Immunological and Antitumor Analysis (LAIA), Department of Antibiotics, Center of Biological Sciences, Federal University of Pernambuco, Cidade Universitária, Av. Prof Artur de Sá, s/n, Recife, PE CEP-50740-525 Brazil
| | | | - Rita de Cássia Pereira de Lima
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Center of Biological Sciences, Federal University of Pernambuco, Cidade Universitária, Av. Prof Moraes Rego, 1235, Recife, PE CEP-50670-901 Brazil
| | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Center of Biological Sciences, Federal University of Pernambuco, Cidade Universitária, Av. Prof Moraes Rego, 1235, Recife, PE CEP-50670-901 Brazil
| | - Aldo Venuti
- HPV-Unit, Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| |
Collapse
|
10
|
Wang Y, Zhang B, Li J, Aipire A, Li Y, Zhang F. Enhanced contraception of canine zona pellucida 3 DNA vaccine via targeting DEC-205 in mice. Theriogenology 2018; 113:56-62. [PMID: 29455062 DOI: 10.1016/j.theriogenology.2018.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 01/24/2018] [Accepted: 02/04/2018] [Indexed: 12/25/2022]
Abstract
Zona pellucida 3 (ZP3) is a potential antigen for the development of contraceptive vaccines to control animal population. In this study, we designed a canine ZP3 (CZP3) DNA vaccine through targeting DEC-205 (named as pcD-scFv-CZP3c) and investigated its contraceptive effect in mice. Female BALB/c mice were intramuscularly immunized 3 times at 2 weeks intervals. After immunization, humoral and cellular immune responses were detected by ELISA and flow cytometry. The results showed that pcD-CZP3 and pcD-scFv-CZP3c induced CZP3-specific antibody (Ab) responses both in serum and vaginal secretions compared to pcDNA3.1. Additionally, compared to pcD-CZP3, pcD-scFv-CZP3c increased the levels of CZP3-specific Abs after a third immunization. Abs induced by these two DNA vaccines could bind with mice and dogs oocytes. Moreover, pcD-scFv-CZP3c enhanced the activation of CD4+ T cells characterized by the increased frequencies of CD4+CD44+ T cells. Finally, the contraceptive effect was evaluated in the immunized mice. These two DNA vaccines significantly decreased a mean litter size of mice compared to pcDNA3.1, but pcD-scFv-CZP3c group showed the smallest mean litter size. The mean litter size of pcD-scFv-CZP3 were 3.2 ± 0.742 and 4.6 ± 1.118 in two mating tests, which were significantly lower than pcDNA3.1(P < 0.001 and P < 0.05). Our results suggest that the CZP3 DNA vaccine targeted with DEC-205 may be a potential strategy for developing a contraceptive DNA vaccine.
Collapse
Affiliation(s)
- Ying Wang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Beibei Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Adila Aipire
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Yijie Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Fuchun Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China.
| |
Collapse
|
11
|
Nair S, Dhodapkar MV. Natural Killer T Cells in Cancer Immunotherapy. Front Immunol 2017; 8:1178. [PMID: 29018445 PMCID: PMC5614937 DOI: 10.3389/fimmu.2017.01178] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/06/2017] [Indexed: 12/27/2022] Open
Abstract
Natural killer T (NKT) cells are specialized CD1d-restricted T cells that recognize lipid antigens. Following stimulation, NKT cells lead to downstream activation of both innate and adaptive immune cells in the tumor microenvironment. This has impelled the development of NKT cell-targeted immunotherapies for treating cancer. In this review, we provide a brief overview of the stimulatory and regulatory functions of NKT cells in tumor immunity as well as highlight preclinical and clinical studies based on NKT cells. Finally, we discuss future perspectives to better harness the potential of NKT cells for cancer therapy.
Collapse
Affiliation(s)
- Shiny Nair
- Yale University, New Haven, CT, United States
| | | |
Collapse
|
12
|
Abstract
Monoclonal antibodies have a variety of applications in research and medicine. Here, we report development of a new method for production of monoclonal antibodies. Our method relies on in vivo RNA transfection rather than peptide vaccination. We took advantage of RNA transcripts complexed with DOTMA and DOPE lipids to transfect mice. Intravenous administration of our RNA vaccine to mice resulted in expression of the antigenic peptides by splenic dendritic cells and detection of the antigens in the serum. The RNA vaccine stimulated production of specific antibodies against the RNA-encoded peptides. We produced monoclonal antibodies against viral, bacterial, and human antigens. In addition, we showed that our RNA vaccine stimulated humoral immunity and rescued mice infected with influenza A virus. Our method could be used as an efficient tool to generate monoclonal antibodies and to stimulate humoral immunity for research and medical purposes.
Collapse
|
13
|
Yuan T, Jiang Y, Li M, Li W. Chronic hepatitis B surface antigen seroclearance-related immune factors. Hepatol Res 2017; 47:49-59. [PMID: 27084584 DOI: 10.1111/hepr.12726] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/01/2016] [Accepted: 04/12/2016] [Indexed: 12/23/2022]
Abstract
The ultimate aims of the treatment of hepatitis B virus infection are the loss of hepatitis B surface antigen (HBsAg) and hepatitis B surface antibody seroconversion. Unfortunately, these goals are rarely reached. Many factors are associated with HBsAg seroconversion, including genetic, immune, and viral factors. However, the mechanism of HBsAg seroclearance, and particularly the immune mechanism, is still difficult to elucidate. The immune factor interferon-α is currently the main antiviral therapy for chronic hepatitis B virus infection. However, a sustained shift from response of HBsAg to hepatitis B surface antibody seroconversion is rarely obtained. Recent studies have revealed that several of the newly identified immune factors are closely related to the removal of HBsAg. In this article, we review recent studies on these immune factors, their influence on hepatitis B progression, and HBsAg seroconversion.
Collapse
Affiliation(s)
- Ting Yuan
- Liver Diseases Research Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongfang Jiang
- Liver Diseases Research Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mei Li
- Liver Diseases Research Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Li
- Liver Diseases Research Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
14
|
Zhang L, Ren J, Zhang H, Cheng G, Xu Y, Yang S, Dong C, Fang D, Zhang J, Yang A. HER2-targeted recombinant protein immuno-caspase-6 effectively induces apoptosis in HER2-overexpressing GBM cells in vitro and in vivo. Oncol Rep 2016; 36:2689-2696. [DOI: 10.3892/or.2016.5088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 07/18/2016] [Indexed: 11/05/2022] Open
|
15
|
Kharkwal SS, Arora P, Porcelli SA. Glycolipid activators of invariant NKT cells as vaccine adjuvants. Immunogenetics 2016; 68:597-610. [PMID: 27377623 DOI: 10.1007/s00251-016-0925-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/22/2016] [Indexed: 11/26/2022]
Abstract
Natural Killer T cells (NKT cells) are a subpopulation of T lymphocytes with unique phenotypic properties and a remarkably broad range of immune effector and regulatory functions. One subset of these cells, known as invariant NKT cells (iNKT cells), has become a significant focus in the search for new and better ways to enhance immunotherapies and vaccination. These unconventional T cells are characterized by their ability to be specifically activated by a range of foreign and self-derived glycolipid antigens presented by CD1d, an MHC class I-related antigen presenting molecule that has evolved to bind and present lipid antigens. The development of synthetic α-galactosylceramides as a family of powerful glycolipid agonists for iNKT cells has led to approaches for augmenting a wide variety of immune responses, including those involved in vaccination against infections and cancers. Here we review the basic background biology of iNKT cells that is relevant to their potential for improving immune responses, and summarize recent work supporting the further development of glycolipid activators of iNKT cells as a new class of vaccine adjuvants.
Collapse
Affiliation(s)
- Shalu Sharma Kharkwal
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Pooja Arora
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Steven A Porcelli
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
16
|
Adjuvant and Novel Treatment of Recurrent Respiratory Papillomatosis. CURRENT OTORHINOLARYNGOLOGY REPORTS 2016. [DOI: 10.1007/s40136-016-0111-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
17
|
Li J, Valentin A, Beach RK, Alicea C, Felber BK, Pavlakis GN. DNA is an efficient booster of dendritic cell-based vaccine. Hum Vaccin Immunother 2016; 11:1927-35. [PMID: 26125100 PMCID: PMC4635890 DOI: 10.1080/21645515.2015.1020265] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
DC-based therapeutic vaccines as a promising strategy against chronic infections and cancer have been validated in several clinical trials. However, DC-based vaccines are complex and require many in vitro manipulations, which makes this a personalized and expensive therapeutic approach. In contrast, DNA-based vaccines have many practical advantages including simplicity, low cost of manufacturing and potent immunogenicity already proven in non-human primates and humans. In this study, we explored whether DC-based vaccines can be simplified by the addition of plasmid DNA as prime or boost to achieve robust CD8-mediated immune responses. We compared the cellular immunity induced in BALB/c and C57BL/6 mice by DC vaccines, loaded either with peptides or optimized SIV Env DNA, and plasmid DNA-based vaccines delivered by electroporation (EP). We found that mature DC loaded with peptides (P-mDC) induced the highest CD8(+) T cell responses in both strains of mice, but those responses were significantly higher in the C57BL/6 model. A heterologous prime-boost strategy (P-DC prime-DNA boost) induced CD8(+) T cell responses similar to those obtained by the P-DC vaccine. Importantly, this strategy elicited robust polyfunctional T cells as well as highest antigen-specific central memory CD8+ T cells in C57BL/6 mice, suggesting long-term memory responses. These results indicate that a DC-based vaccine in combination with DNA in a heterologous DC prime-DNA boost strategy has potential as a repeatedly administered vaccine.
Collapse
Affiliation(s)
- Jinyao Li
- a Human Retrovirus Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute ; Frederick , MD USA
| | | | | | | | | | | |
Collapse
|
18
|
Gableh F, Saeidi M, Hemati S, Hamdi K, Soleimanjahi H, Gorji A, Ghaemi A. Combination of the toll like receptor agonist and α-Galactosylceramide as an efficient adjuvant for cancer vaccine. J Biomed Sci 2016; 23:16. [PMID: 26811064 PMCID: PMC4727273 DOI: 10.1186/s12929-016-0238-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 01/20/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND DNA vaccines have emerged as an attractive approach for the generation of cytotoxic T lymphocytes (CTL). In our previous study, we found That Toll like receptor (TLR) ligands are promising candidates for the development of novel adjuvants for DNA vaccine. To improve the efficacy of DNA vaccine directed against human papillomavirus (HPV) tumors, we evaluated whether co-administration of a TLR4 ligand, monophosphoryl lipid A (MPL), and Natural Killer T Cell Ligand α-Galactosylceramide(α-GalCer) adjuvants with DNA vaccine would influence the anti-tumor efficacy of DNA vaccinations. METHODS We investigated the effectiveness of α-GalCer and MPL combination as an adjuvant with an HPV-16 E7 DNA vaccine to enhance antitumor immune responses. RESULTS By using adjuvant combination for a DNA vaccine, we found that the levels of lymphocyte proliferation, CTL activity, IFN- γ, IL-4 and IL-12 responses, and tumor protection against TC-1 cells were significantly increased compared to the DNA vaccine with individual adjuvants. In addition, inhibition of IL-18 signaling during vaccination decreased IFN-γ responses and tumor protection, and that this inhibition suggested stimulatory role of IL-18 in adjuvant effects of α-GalCer and MPL combination. CONCLUSION The strong adjuvanticity associated with α-GalCer/MPL combination may to be an important tool in the development of novel and strong cancer immunotherapy.
Collapse
Affiliation(s)
- Fateme Gableh
- Infectious Diseases Research Center, Department of Microbiology, Golestan University of Medical Sciences, POBox: 49175-1141, Gorgan, Iran
| | - Mohsen Saeidi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shaghayegh Hemati
- Guilan Science and Research Branch, Islamic Azad University, Rasht, Iran
| | - Kasra Hamdi
- Department of microbiology, Islamic Azad University, Shiraz branch, Shiraz, Iran
| | - Hoorieh Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Tehran, Iran.,Epilepsy Research Center, Institut für Physiologie I, Westfälische Wilhelms-Universität Münster, Robert-Koch-Strasse, Münster, Germany.,Klinik und Poliklinik für Neurochirurgie, Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Amir Ghaemi
- Infectious Diseases Research Center, Department of Microbiology, Golestan University of Medical Sciences, POBox: 49175-1141, Gorgan, Iran. .,Shefa Neuroscience Research Center, Tehran, Iran. .,Department of Virology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
19
|
McKee SJ, Bergot AS, Leggatt GR. Recent progress in vaccination against human papillomavirus-mediated cervical cancer. Rev Med Virol 2015; 25 Suppl 1:54-71. [PMID: 25752816 DOI: 10.1002/rmv.1824] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
It has been more than 7 years since the commercial introduction of highly successful vaccines protecting against high-risk human papillomavirus (HPV) subtypes and the development of cervical cancer. From an immune standpoint, the dependence of cervical cancer on viral infection has meant that HPV proteins can be targeted as strong tumour antigens leading to clearance of the infection and the subsequent protection from cancer. Commercially available vaccines consisting of the L1 capsid protein assembled as virus-like particles (VLPs) induce neutralising antibodies that deny access of the virus to cervical epithelial cells. While greater than 90% efficacy has been demonstrated at the completion of large phase III trials in young women, vaccine developers are now addressing broader issues such as efficacy in boys, longevity of the protection and inducing cross-reactive antibody for oncogenic, non-vaccine HPV strains. For women with existing HPV infection, the prophylactic vaccines provide little protection, and consequently, the need for therapeutic vaccines will continue into the future. Therapeutic vaccines targeting HPVE6 and E7 proteins are actively being pursued with new adjuvants and delivery vectors, combined with an improved knowledge of the tumour microenvironment, showing great promise. This review will focus on recent progress in prophylactic and therapeutic vaccine development and implementation since the publication of end of study data from phase III clinical trials between 2010 and 2012.
Collapse
Affiliation(s)
- Sara J McKee
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | | | | |
Collapse
|
20
|
Unique potential of 4-1BB agonist antibody to promote durable regression of HPV+ tumors when combined with an E6/E7 peptide vaccine. Proc Natl Acad Sci U S A 2015; 112:E5290-9. [PMID: 26351680 DOI: 10.1073/pnas.1514418112] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Antibody modulation of T-cell coinhibitory (e.g., CTLA-4) or costimulatory (e.g., 4-1BB) receptors promotes clinical responses to a variety of cancers. Therapeutic cancer vaccination, in contrast, has produced limited clinical benefit and no curative therapies. The E6 and E7 oncoproteins of human papilloma virus (HPV) drive the majority of genital cancers, and many oropharyngeal tumors. We discovered 15-19 amino acid peptides from HPV-16 E6/E7 for which induction of T-cell immunity correlates with disease-free survival in patients treated for high-grade cervical neoplasia. We report here that intranasal vaccination with these peptides and the adjuvant alpha-galactosylceramide elicits systemic and mucosal T-cell responses leading to reduced HPV(+) TC-1 tumor growth and prolonged survival in mice. We hypothesized that the inability of these T cells to fully reject established tumors resulted from suppression in the tumor microenvironment which could be ameliorated through checkpoint modulation. Combining this E6/E7 peptide vaccine with checkpoint blockade produced only modest benefit; however, coadministration with a 4-1BB agonist antibody promoted durable regression of established genital TC-1 tumors. Relative to other therapies tested, this combination of vaccine and α4-1BB promoted the highest CD8(+) versus regulatory FoxP3(+) T-cell ratios, elicited 2- to 5-fold higher infiltration by E7-specific CTL, and evoked higher densities of highly cytotoxic TcEO (T cytotoxic Eomesodermin) CD8 (>70-fold) and ThEO (T helper Eomesodermin) CD4 (>17-fold) T cells. These findings have immediate clinical relevance both in terms of the direct clinical utility of the vaccine studied and in illustrating the potential of 4-1BB antibody to convert therapeutic E6/E7 vaccines already in clinical trials into curative therapies.
Collapse
|
21
|
Gottschalk C, Mettke E, Kurts C. The Role of Invariant Natural Killer T Cells in Dendritic Cell Licensing, Cross-Priming, and Memory CD8(+) T Cell Generation. Front Immunol 2015; 6:379. [PMID: 26284065 PMCID: PMC4517377 DOI: 10.3389/fimmu.2015.00379] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/11/2015] [Indexed: 12/23/2022] Open
Abstract
New vaccination strategies focus on achieving CD8+ T cell (CTL) immunity rather than on induction of protective antibody responses. While the requirement of CD4+ T (Th) cell help in dendritic cell (DC) activation and licensing, and in CTL memory induction has been described in several disease models, CTL responses may occur in a Th cell help-independent manner. Invariant natural killer T cells (iNKT cells) can substitute for Th cell help and license DC as well. iNKT cells produce a broad spectrum of Th1 and Th2 cytokines, thereby inducing a similar set of costimulatory molecules and cytokines in DC. This form of licensing differs from Th cell help by inducing other chemokines, while Th cell-licensed DCs produce CCR5 ligands, iNKT cell-licensed DCs produce CCL17, which attracts CCR4+ CD8+ T cells for subsequent activation. It has recently been shown that iNKT cells do not only enhance immune responses against bacterial pathogens or parasites but also play a role in viral infections. The inclusion of iNKT cell ligands in influenza virus vaccines enhanced memory CTL generation and protective immunity in a mouse model. This review will focus on the role of iNKT cells in the cross-talk with cross-priming DC and memory CD8+ T cell formation.
Collapse
Affiliation(s)
- Catherine Gottschalk
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University of Bonn , Bonn , Germany
| | - Elisabeth Mettke
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University of Bonn , Bonn , Germany
| | - Christian Kurts
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University of Bonn , Bonn , Germany
| |
Collapse
|
22
|
Carreño LJ, Kharkwal SS, Porcelli SA. Optimizing NKT cell ligands as vaccine adjuvants. Immunotherapy 2015; 6:309-20. [PMID: 24762075 DOI: 10.2217/imt.13.175] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
NKT cells are a subpopulation of T lymphocytes with phenotypic properties of both T and NK cells and a wide range of immune effector properties. In particular, one subset of these cells, known as invariant NKT cells (iNKT cells), has attracted substantial attention because of their ability to be specifically activated by glycolipid antigens presented by a cell surface protein called CD1d. The development of synthetic α-galactosylceramides as a family of powerful glycolipid agonists for iNKT cells has led to approaches for augmenting a wide variety of immune responses, including those involved in vaccination against infections and cancers. Here, we review basic, preclinical and clinical observations supporting approaches to improving immune responses through the use of iNKT cell-activating glycolipids. Results from preclinical animal studies and preliminary clinical studies in humans identify many promising applications for this approach in the development of vaccines and novel immunotherapies.
Collapse
Affiliation(s)
- Leandro J Carreño
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | |
Collapse
|
23
|
Panatto D, Amicizia D, Bragazzi NL, Rizzitelli E, Tramalloni D, Valle I, Gasparini R. Human Papillomavirus Vaccine. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015:231-322. [DOI: 10.1016/bs.apcsb.2015.08.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
24
|
Soong RS, Song L, Trieu J, Knoff J, He L, Tsai YC, Huh W, Chang YN, Cheng WF, Roden RBS, Wu TC, Trimble CL, Hung CF. Toll-like receptor agonist imiquimod facilitates antigen-specific CD8+ T-cell accumulation in the genital tract leading to tumor control through IFNγ. Clin Cancer Res 2014; 20:5456-67. [PMID: 24893628 PMCID: PMC4216740 DOI: 10.1158/1078-0432.ccr-14-0344] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Imiquimod is a Toll-like receptor 7 agonist used topically to treat external genital warts and basal cell carcinoma. We examined the combination of topical imiquimod with intramuscular administration of CRT/E7, a therapeutic human papillomavirus (HPV) vaccine comprised of a naked DNA vector expressing calreticulin fused to HPV16 E7. EXPERIMENTAL DESIGN Using an orthotopic HPV16 E6/E7(+) syngeneic tumor, TC-1, as a model of high-grade cervical/vaginal/vulvar intraepithelial neoplasia, we assessed if combining CRT/E7 vaccination with cervicovaginal deposition of imiquimod could result in synergistic activities promoting immune-mediated tumor clearance. RESULTS Imiquimod induced cervicovaginal accumulation of activated E7-specific CD8(+) T cells elicited by CRT/E7 vaccination. Recruitment was not dependent upon the specificity of the activated CD8(+) T cells, but was significantly reduced in mice lacking the IFNγ receptor. Intravaginal imiquimod deposition induced upregulation of CXCL9 and CXCL10 mRNA expression in the genital tract, which are produced in response to IFNγ receptor signaling and attract cells expressing their ligand, CXCR3. The T cells attracted by imiquimod to the cervicovaginal tract expressed CXCR3 as well as CD49a, an integrin involved in homing and retention of CD8(+) T cells at mucosal sites. Our results indicate that intramuscular CRT/E7 vaccination in conjunction with intravaginal imiquimod deposition recruits antigen-specific CXCR3(+) CD8(+) T cells to the genital tract. CONCLUSIONS Several therapeutic HPV vaccination clinical trials using a spectrum of DNA vaccines, including vaccination in concert with cervical imiquimod, are ongoing. Our study identifies a mechanism by which these strategies could provide therapeutic benefit. Our findings support accumulating evidence that manipulation of the tumor microenvironment can enhance the therapeutic efficacy of strategies that induce tumor-specific T cells.
Collapse
Affiliation(s)
- Ruey-Shyang Soong
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of General Surgery, Chang Gung Memorial Hospital at Keelung, Keelung City, Taiwan. College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Liwen Song
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland. School of Pharmacy, Fudan University, Shanghai, China. Department of Pharmacology and Toxicology, Shanghai Institute of Planned Parenthood Research, Shanghai, China. Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | | | - Jayne Knoff
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Liangmei He
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Ya-Chea Tsai
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Warner Huh
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Wen-Fang Cheng
- Department of Obstetrics and Gynecology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Richard B S Roden
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - T-C Wu
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions, Baltimore, Maryland.
| | - Cornelia L Trimble
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland.
| |
Collapse
|
25
|
Ito H, Ando T, Ando K, Ishikawa T, Saito K, Moriwaki H, Seishima M. Induction of hepatitis B virus surface antigen-specific cytotoxic T lymphocytes can be up-regulated by the inhibition of indoleamine 2, 3-dioxygenase activity. Immunology 2014; 142:614-23. [PMID: 24580128 DOI: 10.1111/imm.12274] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/19/2014] [Accepted: 02/24/2014] [Indexed: 12/12/2022] Open
Abstract
Cytotoxic T lymphocytes (CTLs) are thought to be major effectors involved in viral clearance during acute infections, including hepatitis B virus (HBV) infection. A persistent HBV infection is characterized by a lack of or a weak CTL response to HBV, which may be reflective of tolerance to HBV. Efficient induction of HBV-specific CTLs leads to the clearance of HBV in patients with a chronic HBV infection. Previously, we reported that α-galactosylceramide (α-GalCer), a specific natural killer T (NKT) cell agonist, enhanced the induction of HBV surface antigen (HBsAg)-specific CTLs. In the present study, we found that inhibition of indoleamine 2,3-dioxygenase (IDO) activity enhanced the induction of HBsAg-specific CTLs after immunization with HBsAg and α-GalCer. The administration of HBsAg and α-GalCer increased the production of interleukin-2 and interleukin-12b, which are crucial for the induction of HBsAg-specific CTLs. The production of these cytokines was more strongly enhanced in IDO knockout mice compared with wild-type mice. In addition, α-GalCer induced the production of IDO in CD11b(+) cells, and these cells inhibited proliferation of HBsAg-specific CTLs. Our results lead to strategies for improving the induction of HBsAg-specific CTLs.
Collapse
Affiliation(s)
- Hiroyasu Ito
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Over the past 15 years, investigators have shown that T lymphocytes can recognize not only peptides in the context of MHC class I and class II molecules but also foreign and self-lipids in association with the nonclassical MHC class I-like molecules, CD1 proteins. In this review, we describe the most recent events in the field, with particular emphasis on (a) structural and functional aspects of lipid presentation by CD1 molecules, (b) the development of CD1d-restricted invariant natural killer T (iNKT) cells and transcription factors required for their differentiation, (c) the ability of iNKT cells to modulate innate and adaptive immune responses through their cross talk with lymphoid and myeloid cells, and (d) MR1-restricted and group I (CD1a, CD1b, and CD1c)-restricted T cells.
Collapse
Affiliation(s)
- Mariolina Salio
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DU, United Kingdom;
| | | | | | | |
Collapse
|
27
|
Chen S, Liao C, Lai Y, Fan Y, Lu G, Wang H, Zhang X, Lin MCM, Leng S, Kung HF. De-oncogenic HPV E6/E7 vaccine gets enhanced antigenicity and promotes tumoricidal synergy with cisplatin. Acta Biochim Biophys Sin (Shanghai) 2014; 46:6-14. [PMID: 24240707 DOI: 10.1093/abbs/gmt121] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In order to develop more effective therapeutic vaccines against cancers with high-risk human papillomavirus (HPV) infection, it is crucial to enhance the immunogenicity, eliminate the oncogenicity of oncoproteins, and take a combination of E7- and E6-containing vaccines. It has been shown recently that PE(ΔIII)-E7-KDEL3 (E7), a fusion protein containing the HPV16 oncoprotein E7 and the translocation domain of Pseudomonas aeruginosa exotoxin A, is effective against TC-1 tumor cells inoculated in mice, therefore, we engineered PE(ΔIII)-E6-CRL-KDEL3 (E6), the de-oncogenic versions of the E7 and E6 fusion proteins [i.e. PE(ΔIII)-E7(d)-KDEL3, E7(d), and PE(ΔIII)-E6(d)-CRL-KDEL3, E6(d)] and tested the immunoefficacies of these fusion proteins as mono- and bivalent vaccines. Results indicated that the E7(d) get higher immunogenicity than its wild type and the E6 fusion proteins augmented the immunogenicity and antitumor effects of their E7 counterparts. Furthermore, the bivalent vaccine system E7(d) plus E6(d), in the presence of cisplatin, showed the best tumoristatic and tumoricidal effects against established tumors in vivo. Therefore, it can be concluded that this novel therapeutic vaccine system, upon further optimization, may shed new light on clinical management of HPV-related carcinomas.
Collapse
Affiliation(s)
- Shaochun Chen
- Department of Anatomy and Histoembryology, Kunming Medical University, Kunming 650500, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Amador-Molina A, Hernández-Valencia JF, Lamoyi E, Contreras-Paredes A, Lizano M. Role of innate immunity against human papillomavirus (HPV) infections and effect of adjuvants in promoting specific immune response. Viruses 2013; 5:2624-42. [PMID: 24169630 PMCID: PMC3856406 DOI: 10.3390/v5112624] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 09/30/2013] [Accepted: 10/15/2013] [Indexed: 02/06/2023] Open
Abstract
During the early stages of human papillomavirus (HPV) infections, the innate immune system creates a pro-inflammatory microenvironment by recruiting innate immune cells to eliminate the infected cells, initiating an effective acquired immune response. However, HPV exhibits a wide range of strategies for evading immune-surveillance, generating an anti-inflammatory microenvironment. The administration of new adjuvants, such as TLR (Toll-like receptors) agonists and alpha-galactosylceramide, has been demonstrated to reverse the anti-inflammatory microenvironment by down-regulating a number of adhesion molecules and chemo-attractants and activating keratinocytes, dendritic (DC), Langerhans (LC), natural killer (NK) or natural killer T (NKT) cells; thus, promoting a strong specific cytotoxic T cell response. Therefore, these adjuvants show promise for the treatment of HPV generated lesions and may be useful to elucidate the unknown roles of immune cells in the natural history of HPV infection. This review focuses on HPV immune evasion mechanisms and on the proposed response of the innate immune system, suggesting a role for the surrounding pro-inflammatory microenvironment and the NK and NKT cells in the clearance of HPV infections.
Collapse
Affiliation(s)
- Alfredo Amador-Molina
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, México; E-Mails: (A.A.-M.); (J.F.H.-V.); (A.C.-P.)
| | - José Fernando Hernández-Valencia
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, México; E-Mails: (A.A.-M.); (J.F.H.-V.); (A.C.-P.)
| | - Edmundo Lamoyi
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apartado postal 70228, Ciudad Universitaria, Distrito Federal CP 04510, México; E-Mail:
| | - Adriana Contreras-Paredes
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, México; E-Mails: (A.A.-M.); (J.F.H.-V.); (A.C.-P.)
| | - Marcela Lizano
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, México; E-Mails: (A.A.-M.); (J.F.H.-V.); (A.C.-P.)
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apartado postal 70228, Ciudad Universitaria, Distrito Federal CP 04510, México; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.:+52-55-5573-4662
| |
Collapse
|
29
|
Abstract
Vaccination is the most successful application of immunological principles to human health. Vaccine efficacy needs to be reviewed from time to time and its safety is an overriding consideration. DNA vaccines offer simple yet effective means of inducing broad-based immunity. These vaccines work by allowing the expression of the microbial antigen inside host cells that take up the plasmid. These vaccines function by generating the desired antigen inside the cells, with the advantage that this may facilitate presentation through the major histocompatibility complex. This review article is based on a literature survey and it describes the working and designing strategies of DNA vaccines. Advantages and disadvantages for this type of vaccines have also been explained, together with applications of DNA vaccines. DNA vaccines against cancer, tuberculosis, Edwardsiella tarda, HIV, anthrax, influenza, malaria, dengue, typhoid and other diseases were explored.
Collapse
|
30
|
Lin J, Xu J, Albers AE, Kaufmann AM. New Developments in Therapeutic HPV Vaccines. CURRENT OBSTETRICS AND GYNECOLOGY REPORTS 2012. [DOI: 10.1007/s13669-012-0015-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
31
|
Boosting the Immune Response: The Use of iNKT cell ligands as vaccine adjuvants. ACTA ACUST UNITED AC 2012; 7:436-444. [PMID: 23264781 DOI: 10.1007/s11515-012-1194-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Natural killer T (NKT) cells comprise a small, but important T cell subset and are thought to bridge the innate and adaptive immune responses. The discovery of NKT cells and extensive research on their activating ligands have paved the way for modulation of these potent immunoregulatory cells in order to improve the outcome of various clinical conditions. Efforts to modulate NKT cell effector functions have ranged from therapy for influenza to anti-tumor immunotherapy. These approaches have also led to the use of NKT cell agonists such as α-Galactosylceramide (α-GalCer) and its analogs as vaccine adjuvants, an approach that is aimed at boosting specific B and T cell responses to a vaccine candidate by concomitant activation of NKT cells. In this review we will provide a comprehensive overview of the efforts made in using α-GalCer and its analogs as vaccine adjuvants. The diverse array of vaccination strategies used, as well as the role of NKT cell activating adjuvants will be discussed, with focus on vaccines against malaria, HIV, influenza and tumor vaccines. Collectively, these studies demonstrate the efficacy of NKT cell-specific agonists as adjuvants and further suggest that these compounds warrant serious consideration during the development of vaccination strategies.
Collapse
|
32
|
Bergot AS, Kassianos A, Frazer IH, Mittal D. New Approaches to Immunotherapy for HPV Associated Cancers. Cancers (Basel) 2011; 3:3461-95. [PMID: 24212964 PMCID: PMC3759206 DOI: 10.3390/cancers3033461] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 08/26/2011] [Accepted: 08/29/2011] [Indexed: 02/08/2023] Open
Abstract
Cervical cancer is the second most common cancer of women worldwide and is the first cancer shown to be entirely induced by a virus, the human papillomavirus (HPV, major oncogenic genotypes HPV-16 and -18). Two recently developed prophylactic cervical cancer vaccines, using virus-like particles (VLP) technology, have the potential to prevent a large proportion of cervical cancer associated with HPV infection and to ensure long-term protection. However, prophylactic HPV vaccines do not have therapeutic effects against pre-existing HPV infections and do not prevent their progression to HPV-associated malignancy. In animal models, therapeutic vaccines for persisting HPV infection can eliminate transplantable tumors expressing HPV antigens, but are of limited efficacy in inducing rejection of skin grafts expressing the same antigens. In humans, clinical trials have reported successful immunotherapy of HPV lesions, providing hope and further interest. This review discusses possible new approaches to immunotherapy for HPV associated cancer, based on recent advances in our knowledge of the immunobiology of HPV infection, of epithelial immunology and of immunoregulation, with a brief overview on previous and current HPV vaccine clinical trials.
Collapse
Affiliation(s)
- Anne-Sophie Bergot
- Author to whom correspondence should be addressed; E-Mails: (A.-S.B); (D.M.); Tel.: +61 (07) 3176 2769; Fax: +61 7 3176 5946
| | | | | | - Deepak Mittal
- Author to whom correspondence should be addressed; E-Mails: (A.-S.B); (D.M.); Tel.: +61 (07) 3176 2769; Fax: +61 7 3176 5946
| |
Collapse
|