1
|
Emadi M, Jahanshiri F, Kaveh K, Hair-Bejo M, Ideris A, Alimon R. Tryptophan Stimulates Immune Response in Broiler Chickens Challenged with Infectious Bursal Disease Vaccine. ACTA ACUST UNITED AC 2010. [DOI: 10.3923/javaa.2010.610.616] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
2
|
Emadi M, Jahanshiri F, Jalalian FA, Kaveh K, Bejo M, Ideris A, Assumaidae A, Alimon R. Immunostimulatory Effects of Arginine in Broiler Chickens Challenged with Vaccine Strain of Infectious Bursal Disease Virus. ACTA ACUST UNITED AC 2010. [DOI: 10.3923/javaa.2010.594.600] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
3
|
Hendrickson RC, Cicinnati VR, Albers A, Dworacki G, Gambotto A, Pagliano O, Tüting T, Mayordomo JI, Visus C, Appella E, Shabanowitz J, Hunt DF, DeLeo AB. Identification of a 17beta-hydroxysteroid dehydrogenase type 12 pseudogene as the source of a highly restricted BALB/c Meth A tumor rejection peptide. Cancer Immunol Immunother 2010; 59:113-24. [PMID: 19562340 PMCID: PMC2855844 DOI: 10.1007/s00262-009-0730-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 06/03/2009] [Indexed: 12/23/2022]
Abstract
Mass spectrometric analysis identified the peptide recognized by a cytotoxic T lymphocyte (CTL) specific for the chemically induced BALB/c Meth A sarcoma as derived from a 17beta-hydroxysteroid dehydrogenase type 12 (Hsd17b12) pseudogene present in the BALB/c genome, but only expressed in Meth A sarcoma. The sequence of the peptide is TYDKIKTGL and corresponds to Hsd17b12(114-122) with threonine instead of isoleucine at codon 114 and is designated Hsd17b12(114T). Immunization of mice with an Hsd17b12(114T) peptide-pulsed dendritic cell-based vaccine or a non-viral plasmid construct expressing the Hsd17b12(114T) peptide protected the mice from lethal Meth A tumor challenge in tumor rejection assays. A Hsd17b12(114-122) peptide-pulsed vaccine was ineffective in inducing resistance in mice to Meth A sarcoma. These results confirm the immunogenicity of the identified tumor peptide, as well as demonstrate the efficacies of these vaccine vehicles. These findings suggest that the role of the human homolog of Hsd17b12, HSD17B12, as a potential human tumor antigen be explored.
Collapse
Affiliation(s)
| | - Vito R. Cicinnati
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Andreas Albers
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Grzegorz Dworacki
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Andrea Gambotto
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Ornella Pagliano
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Thomas Tüting
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Jose I. Mayordomo
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Carmen Visus
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Ettore Appella
- Laboratory of Cell Biology, National Cancer Institute, Bethesda, MD 20892 USA
| | - Jeffrey Shabanowitz
- Department of Chemistry, University of Virginia, Charlottesville, VA, 22901 USA
| | - Donald F. Hunt
- Department of Chemistry, University of Virginia, Charlottesville, VA, 22901 USA
- Department of Pathology, University of Virginia, Charlottesville, VA, 22901 USA
| | - Albert B. DeLeo
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Division of Basic Research, Hillman Cancer Center, Research Pavilion, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| |
Collapse
|
4
|
DeLeo AB, Whiteside TL. Development of multi-epitope vaccines targeting wild-type sequence p53 peptides. Expert Rev Vaccines 2008; 7:1031-40. [PMID: 18767952 DOI: 10.1586/14760584.7.7.1031] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Loss of p53 tumor-suppressor function is the most common abnormality in human cancer, which can result in enhanced presentation to immune cells of wild-type (wt)-sequence peptides from tumor p53 molecules, thus providing the rationale for wt p53 peptide-based cancer vaccines. We review evidence from preclinical murine tumor models and preclinical studies that led to the clinical introduction of wt p53 peptide-based vaccines for cancer immunotherapy. Overall, this review illustrates the complex process of wt p53 epitope selection and the issues and concerns involved in the application of p53-based vaccines for patients with cancer.
Collapse
Affiliation(s)
- Albert B DeLeo
- Department of Pathology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | | |
Collapse
|
5
|
Justesen S, Buus S, Claesson MH, Pedersen AE. Addition of TAT protein transduction domain and GrpE to human p53 provides soluble fusion proteins that can be transduced into dendritic cells and elicit p53-specific T-cell responses in HLA-A*0201 transgenic mice. Immunology 2007; 122:326-34. [PMID: 17610503 PMCID: PMC2266015 DOI: 10.1111/j.1365-2567.2007.02643.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The protein p53 has been shown to be an efficient tumour antigen in both murine and human cancer vaccine studies and cancer vaccines targeting p53 based on major histocompatibility complex (MHC) class I binding p53-derived peptides that induce cytotoxic T lymphocytes (CTLs) without p53-specific CD4(+) T-cell help have been tested by several research groups including ours. To obtain such CD4(+) T-cell help and cover a broader repertoire of MHC haplotypes we have previously attempted to produce recombinant human p53 for vaccination purposes. However, attempts to refold a hexahis-tagged p53 protein in our laboratory were unsuccessful. Here, we show that fusion of an 11-amino-acid region of the human immunodeficiency virus TAT protein transduction domain (PTD) to human p53 increases the solubility of the otherwise insoluble p53 protein and this rTAT-p53 protein can be transduced into human monocyte-derived dendritic cells (DCs). The induction of a p53-specific HLA-A*0201 immune response was tested in HLA-A*0201/K(b) transgenic mice after immunization with rTAT-p53-transduced bone-marrow-derived DCs. In these mice, p53-specific CD4(+) and CD8(+) T-cell proliferation was observed and immunization resulted in the induction of HLA-A*0201-restricted CTLs specific for two human p53-derived HLA-A*0201-binding peptides, p53(65-73) and p53(149-157). Addition of GrpE to generate rTAT-GrpE-p53 led to a further increase in protein solubility and to a small increase in DC maturation but did not increase the observed p53-specific T-cell responses. The use of rTAT-p53 in ongoing clinical protocols should be applicable and offers advantages to current strategies omitting the use of HLA-typed patients.
Collapse
Affiliation(s)
- S Justesen
- Institute of International Health, Immunology and Microbiology, The Panum Institute, University of Copenhagen, Denmark
| | | | | | | |
Collapse
|
6
|
Yan J, Liu X, Wang Y, Jiang X, Liu H, Wang M, Zhu X, Wu M, Tien P. Enhancing the potency of HBV DNA vaccines using fusion genes of HBV-specific antigens and the N-terminal fragment of gp96. J Gene Med 2007; 9:107-21. [PMID: 17256801 DOI: 10.1002/jgm.998] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Many clinical trials show that DNA vaccine potency needs to be greatly enhanced. We have reported that the N-terminal fragment of glycoprotein 96 (gp96) is able to produce an adjuvant effect for production of cytotoxic T-lymphocytes (CTLs) with hepatitis B virus (HBV)-specific peptides. Here, we report a new strategy for HBV DNA vaccine design using a partial gp96 sequence. MATERIALS AND METHODS We linked the N-terminal 1-355aa (N355) of gp96 to HBV genes encoding for structural proteins, the major S and middle S2S envelope proteins and the truncated core HBcAg (1-149aa). ELISPOT, tetramer staining and intracellular IFN-gamma assay were performed to analyze the induced cellular immune responses of our DNA constructs in BALB/c mice and HLA-A2 transgenic mice. The relative humoral immune responses were analyzed in different IgG isotypes. RESULTS The fusion genes induced 2- to 6-fold higher HBV-specific CD8(+) T cells as compared to the antigens alone. There was an approximate 10-fold decrease in the humoral immune responses with fusion genes based on HBV envelope proteins. Interestingly, the decreased humoral immune responses were not observed when antigens and plasmid encoding N355 were co-delivered. However, an approximate 20-fold higher antibody level was induced when linking N355 to a truncated HBcAg. Immunization by intramuscular injection resulted in predominantly IgG2a antibodies, which indicated that these vaccines preferentially prime Th1 responses. CONCLUSIONS We constructed highly immunogenic fusions by linking the N-terminal fragment of gp96 to HBV antigens. Our results imply that the N-terminal fragment of gp96 may be used as a molecular adjuvant to enhance the potency of DNA vaccines.
Collapse
Affiliation(s)
- Jiabin Yan
- Center for Molecular Virology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100080, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Hsieh MK, Wu CC, Lin TL. The effect of co-administration of DNA carrying chicken interferon-γ gene on protection of chickens against infectious bursal disease by DNA-mediated vaccination. Vaccine 2006; 24:6955-65. [PMID: 17050043 DOI: 10.1016/j.vaccine.2006.05.127] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Revised: 04/26/2006] [Accepted: 05/02/2006] [Indexed: 11/24/2022]
Abstract
The purpose of the present study was to determine whether DNA vaccination by co-administration of DNA coding for chicken interferon-gamma (IFN-gamma) gene and DNA encoding for the VP243 gene of IBDV could enhance immune response and protection efficacy of chickens against challenge by IBDV. Plasmids carrying VP243 gene of IBDV strain variant E (VE) (P/VP243/E) and chicken IFN-gamma gene (P/cIFN-gamma) were constructed, respectively. One-day-old chickens were intramuscularly injected with P/VP243/E, or P/cIFN-gamma, or both once, twice, or three times into the thigh muscle of one leg or the thigh muscles of two separate legs at weekly intervals. Chickens were orally challenged with IBDV strain VE at 3 weeks of age and observed for 10 days. Chickens receiving two plasmids in the same site two times had significantly higher (P<0.05) bursal lesion scores and significantly lower (P<0.05) bursa weight/body weight ratios than those that only received P/VP243/E two or three times. Chickens inoculated with two plasmids separately in the thigh muscles of different legs or P/VP243/E two times had 33-50% protection and those receiving two plasmids in the same sites did not have any protection against IBD. The enzyme-linked immunosorbent assay (ELISA) and virus neutralization (VN) titers to IBDV of chickens in the groups with three doses of P/VP243/E were significantly higher (P<0.05) than those in groups receiving two doses of P/VP243/E or P/VP243/E and P/cIFN-gamma. Chickens protected by DNA vaccination did not have detectable IBDV antigen in the bursae as determined by immunofluorescent antibody assay (IFA). The results indicated that co-administration of plasmid encoding chicken IFN-gamma gene with plasmid encoding a large segment gene of the IBDV did not enhance immune response and protection against challenge by IBDV.
Collapse
Affiliation(s)
- Ming Kun Hsieh
- Department of Veterinary Pathobiology, School of Veterinary Medicine, Purdue University, West Lafayette, IN 47907-2065, USA
| | | | | |
Collapse
|
8
|
Abstract
Significant progress made in the field of tumor immunology by the characterization of a large number of tumor antigens, and the better understanding of the mechanisms preventing immune responses to malignancies has led to the extensive study of cancer immunization approaches such as DNA vaccines encoding tumor antigens. This article reviews major aspects of DNA immunization in cancer. It gives a brief history and then discusses the proposed mechanism of action, preclinical and clinical studies, and methods of enhancing the immune responses induced by DNA vaccines.
Collapse
Affiliation(s)
- Rodica Stan
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | |
Collapse
|
9
|
U'Ren L, Kedl R, Dow S. Vaccination with liposome--DNA complexes elicits enhanced antitumor immunity. Cancer Gene Ther 2006; 13:1033-44. [PMID: 16841080 DOI: 10.1038/sj.cgt.7700982] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cationic liposomes have been shown to potentiate markedly the ability of plasmid DNA to activate innate immune responses. We reasoned therefore that liposome-DNA complexes (LDC) could be used to produce more effective plasmid DNA vaccines for cancer. To test this hypothesis, tumor-bearing mice were vaccinated with conventional plasmid DNA vaccines or with LDC vaccines encoding model tumor antigens and CD8(+) T-cell responses and antitumor activity were assessed. We found that although plasmid DNA vaccines generated large increases in antigen-specific CD8(+) T cells, they failed to elicit significant antitumor immunity. In contrast, LDC vaccines elicited large numbers of antigen-specific CD8(+) T cells and also generated significant antitumor activity against established tumors. The antitumor activity elicited by immunization with LDC vaccines was mediated primarily by CD8(+) T cells. Studies of the interaction of LDC with antigen-presenting cells found that LDC triggered dendritic cell production of interleukin-12 and interferon (IFN)-gamma production by natural killer cells in vivo. Activation by LDC was also accompanied by upregulation of costimulatory molecule expression. These findings suggest that by concurrently activating strong systemic innate immune responses and generating cytotoxic T-lymphocyte responses, LDC may be used to increase the effectiveness of therapeutic plasmid DNA vaccination for cancer.
Collapse
Affiliation(s)
- L U'Ren
- Department of Microbiology, Colorado State University, Ft Collins, CO 80523, USA
| | | | | |
Collapse
|
10
|
Steitz J, Wenzel J, Gaffal E, Tüting T. Initiation and regulation of CD8+T cells recognizing melanocytic antigens in the epidermis: implications for the pathophysiology of vitiligo. Eur J Cell Biol 2005; 83:797-803. [PMID: 15679123 DOI: 10.1078/0171-9335-00423] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Antigen-specific CD8+T lymphocytes play an important role in defense against cutaneous microbial infection and skin cancer as well as in the pathophysiology of autoimmune skin disease such as lupus erythematodes and vitiligo. We have explored the role of CD8+ cytotoxic T lymphocytes (CTL) in an experimental mouse model of vitiligo, a pigmentation disorder characterized by focal loss of melanocytes in the skin. Using genetic immunization techniques we found that pigment cells in the epidermis can be destroyed by CD8+ T cells specifically recognizing a single H2-Kb-binding peptide derived from the model melanocytic self antigen tyrosinase-related protein 2 (TRP2), a melanosomal enzyme involved in pigment synthesis. Experimental evidence suggests that peripheral tolerance of pigment cell-specific cytotoxic CD8+T cells is regulated in two steps. In the induction phase, stimulation and expansion of these T cells in vivo strictly depends on CD4+ T cell help. In the effector phase, autoimmune destruction of melanocytes in the skin depends on local inflammation facilitating the migration of T cells into the epidermis and supporting effector functions. Our results suggest that accidental stimulation of CD8+ CTL recognizing MHC class I-binding peptides derived from melanocytic proteins in the context of an inflammatory skin disease may play an important role in the pathophysiology of vitiligo. Further investigations will address the role of chemokines, chemokine receptors and adhesion molecules in this experimental system and will reveal the role of keratinocytes and Langerhans cells in regulating cutaneous CD8+ T cell responses.
Collapse
Affiliation(s)
- Julia Steitz
- Laboratory of Experimental Dermatology, Department of Dermatology, University of Bonn, Bonn, Germany
| | | | | | | |
Collapse
|
11
|
Cicinnati VR, Dworacki G, Albers A, Beckebaum S, Tüting T, Kaczmarek E, DeLeo AB. Impact of p53-based immunization on primary chemically-induced tumors. Int J Cancer 2004; 113:961-70. [PMID: 15514940 DOI: 10.1002/ijc.20686] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In mice as well as humans, cytotoxic T lymphocytes (CTL) specific for wild-type-sequence (wt) p53 peptides have been shown to react against a wide range of tumors, but not normal cells. As such, they are attractive candidates for developing broadly applicable cancer vaccines. Of particular interest is the potential of using p53-based vaccines in high-risk individuals to prevent cancer. Methylcholanthrene, an immunosuppressive polycyclic hydrocarbon carcinogen implicated as a causative agent in human cancers, has long been used to induce murine tumors with a high incidence of genetic alterations and sensitivity to wt p53-specific CTL. To analyze the potential of p53-based vaccines on primary tumors, we evaluated the efficacy of DNA and dendritic cell vaccines targeting wt p53 peptides given to methylcholanthrene-treated mice in the protection or therapy settings. The results indicate that the efficacy of these vaccines relative to reducing tumor incidence were severely compromised by vaccine-induced tumor escape. As compared to tumors induced in non-immunized mice, a higher incidence of epitope-loss tumors was detected in tumors from the immunized mice. The increase in tumor escape arose as a consequence of either increased frequencies of mutations within/flanking p53 epitope-coding regions or downregulation of expression of the major histocompatibility complex Class I molecules that present these epitopes for T cell recognition These findings are consistent with current views of immunoselection occurring in patients receiving tumor peptide-based immunotherapy, and impact on the design and implementation of p53-based vaccines, in particular, those aimed at treating individuals at high risk for developing cancer.
Collapse
Affiliation(s)
- Vito R Cicinnati
- Division of Basic Research, University of Pittsburgh Cancer Institute, Department of Pathology, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Keyser J, Schultz J, Ladell K, Elzaouk L, Heinzerling L, Pavlovic J, Moelling K. IP-10-encoding plasmid DNA therapy exhibits anti-tumor and anti-metastatic efficiency. Exp Dermatol 2004; 13:380-90. [PMID: 15186325 DOI: 10.1111/j.0906-6705.2004.00191.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We report here that the interferon-induced protein of 10 kDa (IP-10 or CXCL10) elicits strong anti-tumor and anti-metastatic responses in mice when administered by plasmid DNA. Intratumoral but not intramuscular IP-10 DNA inoculation resulted in reduced tumor formation of malignant melanoma (B16F10) and Lewis lung carcinoma (LL/2) in C57BL/6 mice. In addition, plasmid DNA-encoding IP-10 substantially reduced the establishment of metastases when injected systemically by the intramuscular route. In contrast to the primary tumor model, the anti-metastatic effect of DNA-encoding IP-10 was primarily mediated by NK cells. Compared to DNA-encoding interleukin-12 (IL-12), therapy with DNA-encoding IP-10 exhibits lower efficacy against primary melanoma tumors but equivalent efficacy against primary Lewis lung tumors and against B16F10 lung metastasis formation. Co-administration of DNA-encoding IP-10 and IL-12 enhanced the anti-tumor activity of IL-12 in the lung metastasis model but had little effect in the local treatment of established subcutaneous tumors. Interestingly, treatment of nude mice lacking T lymphocytes with DNA-encoding IP-10 or IL-12 still resulted in a pronounced reduction of tumor growth or metastasis formation.
Collapse
Affiliation(s)
- Johanna Keyser
- Institute of Medical Virology, University of Zurich, Gloriastr., Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
13
|
Sudowe S, Ludwig-Portugall I, Montermann E, Ross R, Reske-Kunz AB. Transcriptional targeting of dendritic cells in gene gun-mediated DNA immunization favors the induction of type 1 immune responses. Mol Ther 2003; 8:567-75. [PMID: 14529829 DOI: 10.1016/s1525-0016(03)00242-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cutaneous dendritic cells (DC) are pivotal for the elicitation of antigen-specific immune responses following gene gun-mediated biolistic transfection of the skin. We transcriptionally targeted transgene expression to DC using vectors containing the murine fascin promoter (pFascin) to control antigen production and compared the immune response elicited with conventional DNA immunization using plasmid constructs with the ubiquitously active CMV promoter (pCMV). Biolistic transfection with pFascin initiated a marked type 1 immune response characterized by the occurrence of a large population of IFN-gamma-producing T helper (Th) cells in spleen and draining lymph nodes. Consistently, immunoglobulin production was dominated by IgG2a antibodies. In contrast, the humoral response after repeated administration of pCMV was strongly enhanced and characterized by a type 2-like isotype pattern (IgG1 > IgG2a). Cytokine production analysis in vitro indicated compartmentalization of the immune response, revealing large numbers of IL-4-producing Th cells in the lymph nodes and dominant presence of IFN-gamma-producing Th cells in the spleen. Biolistic transfection with pFascin, like immunization with pCMV, led to potent induction of cytotoxic T cells as was assessed by JAM test. Thus gene gun immunization with plasmids that focus transgene expression and antigen production specifically to DC propagates type 1-biased cellular immune responses.
Collapse
Affiliation(s)
- Stephan Sudowe
- Clinical Research Unit Allergology, Department of Dermatology, Johannes Gutenberg-University, Obere Zahlbacher Strasse 63, D-55101 Mainz, Germany.
| | | | | | | | | |
Collapse
|
14
|
Frelin L, Alheim M, Chen A, Söderholm J, Rozell B, Barnfield C, Liljeström P, Sällberg M. Low dose and gene gun immunization with a hepatitis C virus nonstructural (NS) 3 DNA-based vaccine containing NS4A inhibit NS3/4A-expressing tumors in vivo. Gene Ther 2003; 10:686-99. [PMID: 12692597 DOI: 10.1038/sj.gt.3301933] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The hepatitis C virus (HCV) protease and helicase encompasses the nonstructural (NS) 3 protein and the cofactor NS4A, which targets the NS3/4A-complex to intracellular membranes. We here evaluate the importance of NS4A in NS3-based genetic immunogens. A full-length genotype 1 NS3/4A gene was cloned into a eucaryotic expression vector in the form of NS3/4A and NS3 alone. Transient transfections revealed that the inclusion of NS4A increased the expression levels of NS3. Subsequently, immunization with the NS3/4A gene primed 10- to 100-fold higher levels of NS3-specific antibodies as compared to immunization with the NS3 gene. Humoral responses primed by the NS3/4A gene had a higher IgG2a/IgG1 ratio (>20) as compared to the NS3 gene (3.0), suggesting a T helper 1-skewed response. Low dose i.m. (10 microg) immunization with the NS3/4A gene inhibited the growth of NS3/4A-expressing tumor cells in vivo, whereas the NS3 gene alone or NS3 protein did not. We then evaluated the efficiency of the NS3/4A gene administered by the gene gun, at the same doses used for humans, in priming cytotoxic T lymphocyte (CTL) responses. Three to four 4 microg doses of the NS3/4A gene primed CTL at a precursor frequency of 2-4%, which inhibited the growth of NS3/4A-expressing tumor cells in vivo. Thus, NS4A enhances the expression levels and immunogenicity of NS3, and an NS3/4A gene delivered transdermally could be a therapeutic vaccine candidate.
Collapse
Affiliation(s)
- L Frelin
- Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Björck P, Lie WR, Woulfe SL, Klein BK, Olson W, Storkus WJ. Progenipoietin-generated dendritic cells exhibit anti-tumor efficacy in a therapeutic murine tumor model. Int J Cancer 2002; 100:586-91. [PMID: 12124809 DOI: 10.1002/ijc.10529] [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] [Indexed: 11/05/2022]
Abstract
Progenipoietin (ProGP-4) is a chimeric molecule, exhibiting both Flt-3 and granulocyte-colony stimulating factor (G-CSF) receptor agonist activities. Subcutaneous administration of ProGP-4 to BALB/c mice at a dose of 40-100 microg/day for up to 12 consecutive days induces both CD11c(+) dendritic cells (DCs) and CD11c(-)/CD11b(+) granulocytes in spleen, blood and lymph nodes of treated animals. Peak numbers of all cell populations were observed on day 7 of treatment, with CD11c(+) DCs representing approximately 8% of total splenocytes at that time. Approximately 40-50% of these CD11c(+) cells were also able to endocytose and process the exogenous fluorescent antigen DQ-BSA. As a test of their therapeutic utility, freshly prepared CD11c(+) DCs were pulsed with a defined tumor-associated peptide epitope (murine p53(232-240)) and injected as a vaccine into BALB/c mice bearing day 7 established CMS4 sarcomas. Similarly prepared DCs were injected again 1 week later. Based on our results, we conclude that (i) both peptide-pulsed CD11c(+) DCs (harvested directly from ProGP-4 treated mice) and pulsed bone marrow-derived DCs effectively slow the growth of or mediate the regression (in 25 of 89 [28%] cases) of CMS4 tumors, and (ii) nonpulsed DCs mediated minimal or no therapeutic effect. These data support the ability of ProGP-4 to enhance the peripheral frequencies of DCs that exhibit therapeutic efficacy when applied as a vaccine to treat tumor-bearing animals.
Collapse
Affiliation(s)
- Pia Björck
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Egan MA, Israel ZR. The use of cytokines and chemokines as genetic adjuvants for plasmid DNA vaccines. ACTA ACUST UNITED AC 2002. [DOI: 10.1016/s1529-1049(02)00051-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
17
|
Sudowe S, Montermann E, Steitz J, Tüting T, Knop J, Reske-Kunz AB. Efficacy of recombinant adenovirus as vector for allergen gene therapy in a mouse model of type I allergy. Gene Ther 2002; 9:147-56. [PMID: 11857073 DOI: 10.1038/sj.gt.3301625] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2001] [Accepted: 11/29/2001] [Indexed: 11/09/2022]
Abstract
DNA-based immunization represents an attractive alternative approach to the current treatment of allergic diseases by specific immunotherapy with allergen extracts. In this study, we used a replication-deficient adenovirus vector (AdCMV), to examine the in vivo efficacy of preventive and therapeutic genetic immunization in a mouse model of type I allergy. Primary immunization with a recombinant adenovirus expressing the model antigen beta-galactosidase (AdCMV-(beta)gal) induced a Th1 immune response (predominance of IgG2a antibodies, high frequency of IFN-gamma producing T cells) and large numbers of cytotoxic T lymphocytes. Prophylactic vaccination with AdCMV-(beta)gal abolished the production of specific IgE following subsequent immunization with (beta)gal-protein, and skewed the Th2-biased immune response to a Th1-orientated response. In contrast, therapeutic administration of AdCMV-(beta)gal after priming with (beta)gal-protein neither significantly inhibited ongoing IgE production nor modulated a manifest Th2 immune response. Thus, allergen gene transfer via recombinant adenovirus represents an effective method to establish protection against the development of allergic disorders, but does not qualify as a therapeutic tool to interfere with ongoing high IgE production.
Collapse
Affiliation(s)
- S Sudowe
- Clinical Research Unit Allergology, Department of Dermatology, Johannes Gutenberg-University, Mainz, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Deng H, Kowalczyk D, O I, Blaszczyk-Thurin M, Quan Xiang Z, Giles-Davis W, Ertl HCJ. A modified DNA vaccine to p53 induces protective immunity to challenge with a chemically induced sarcoma cell line. Cell Immunol 2002; 215:20-31. [PMID: 12142033 DOI: 10.1016/s0008-8749(02)00004-7] [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] [Indexed: 10/27/2022]
Abstract
Different vaccine constructs based on DNA vaccines and viral recombinant vaccines expressing mouse p53 were compared for induction of protective immune responses to challenge with a sarcoma cell line that expresses high levels of mutated p53 protein. Viral recombinant vaccines based on E1-deleted adenovirus or vaccinia virus recombinants expressing p53 with wild-type sequences in the mutational hotspot domain and a single mutation in the tetramerization domain (p53(mu338)) failed to induce protection against progression of this tumor cell line. A DNA vaccine expressing a form of p53 carrying the same point mutations as the tumor cell line showed low efficacy that was comparable to that of a DNA vaccine expressing p53(mu338). Efficacy of the DNA vaccine was augmented upon expressing p53(mu338) as a fusion protein linked to a viral leader sequence. Other modifications such as fusion to the signal sequence of the lysosome-associated membrane protein (LAMP) or ubiquitin failed to improve the efficacy of the vaccine to p53. Protection mediated by CD4(+) and CD8(+) T cells was specific for p53.
Collapse
Affiliation(s)
- Honying Deng
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Creusot RJ, Thomsen LL, van Wely CA, Topley P, Tite JP, Chain BM. Early commitment of adoptively transferred CD4+ T cells following particle-mediated DNA vaccination: implications for the study of immunomodulation. Vaccine 2001; 19:1678-87. [PMID: 11166891 DOI: 10.1016/s0264-410x(00)00405-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The early responses of CD4+ T cells to particle-mediated DNA immunisation were investigated using OVA-specific TCR-transgenic CD4+ T cells. Following adoptive transfer of these cells, mice were immunised by delivery into the skin of a plasmid encoding ovalbumin. Transgenic T cells underwent a rapid and transient antigen-specific activation, followed by clonal expansion (up to approximately 6% of total lymphocytes). Immunisation with ovalbumin in CFA evoked similar responses with slightly faster kinetics. Numerous antigen-specific T cells synthesising IFN-gamma (Th1) and IL-4 (Th2) were detectable using both intracellular staining and ELISPOT assays. This study provides a quantitative analysis of both T cell proliferation and Th1/Th2 balance following particle-mediated DNA immunisation and establishes a robust and sensitive model in which to assess modulation of helper T cell responses in DNA vaccination.
Collapse
Affiliation(s)
- R J Creusot
- Department of Immunology, Windeyer Institute of Medical Sciences, University College London, 46 Cleveland Street, W1P 6DB, London, UK
| | | | | | | | | | | |
Collapse
|
20
|
Gambotto A, Dworacki G, Cicinnati V, Kenniston T, Steitz J, Tüting T, Robbins PD, DeLeo AB. Immunogenicity of enhanced green fluorescent protein (EGFP) in BALB/c mice: identification of an H2-Kd-restricted CTL epitope. Gene Ther 2000; 7:2036-40. [PMID: 11175316 DOI: 10.1038/sj.gt.3301335] [Citation(s) in RCA: 149] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Enhanced green fluorescent protein (EGFP) is a novel marker gene product, which is readily detectable using techniques of fluorescence microscopy, flow cytometry, or macroscopic imaging. In the present studies, we have examined the immunogenicity of EGFP in murine models. A stable transfectant of the transplantable CMS4 sarcoma of BALB/c origin expressing EGFP, CMS4-EGFP-Zeo, was generated. Splenocytes harvested from mice immunized with a recombinant adenovirus expressing EGFP (Ad-EGFP) were restimulated in vitro with CMS4-EGFP-Zeo. Effector lymphocytes displayed strong cytotoxicity against CMS4-EGFP-Zeo, but not against mock-transfected CMS4-Zeo tumor cells. A number of candidate H2-Kd-binding peptides derived from the EGFP protein were chosen according to an epitope prediction program and synthesized. These peptides were tested for their ability to bind to H2-Kd molecules and stimulate IFNgamma-production by splenocytes harvested from Ad-EGFP-immunized mice. Using this methodology, the peptide, HYLSTQSAL (corresponding to EGFP200-208) which strongly binds to H2-Kd molecules, was identified as a naturally occurring epitope of EGFP. These results should facilitate the use of EGFP as a model tumor antigen in BALB/c mice.
Collapse
Affiliation(s)
- A Gambotto
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Steitz J, Brück J, Steinbrink K, Enk A, Knop J, Tüting T. Genetic immunization of mice with human tyrosinase-related protein 2: implications for the immunotherapy of melanoma. Int J Cancer 2000; 86:89-94. [PMID: 10728600 DOI: 10.1002/(sici)1097-0215(20000401)86:1<89::aid-ijc14>3.0.co;2-i] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The melanosomal protein TRP2 expressed by melanocytes and most melanoma cells is an attractive, clinically relevant model antigen for the experimental development of melanoma immunotherapy in mice. A peptide shared by murine and human TRP2 can be recognized by melanoma-reactive CTL in C57BL/6 mice, as well as in human melanoma patients. Previous experiments demonstrated that gene gun immunization of mice with plasmid DNA encoding autologous murine TRP2 was unable to induce protective immunity against B16 melanoma cells naturally expressing TRP2. In the present study, we investigated whether the use of cDNA encoding xenogeneic human TRP2, which is highly homologous to murine TRP2, would be more effective. Genetic immunization of mice with human TRP2 resulted in coat depigmentation as a sign of autoimmune-mediated destruction of melanocytes and provided significant protection against metastatic growth of B16 melanoma Induction of protective immunity was associated with TRP2-reactive antibodies and CD8+ T cells. Furthermore, immunization with recombinant adenovirus was more effective than immunization with plasmid DNA using the gene gun. Our results provide new insights for the development of antigen-specific immunotherapy of melanoma.
Collapse
Affiliation(s)
- J Steitz
- Department of Dermatology, J. Gutenberg-University, Mainz, Germany
| | | | | | | | | | | |
Collapse
|
22
|
Strong TV. Gene therapy for carcinoma of the breast: Genetic immunotherapy. Breast Cancer Res 2000; 2:15-21. [PMID: 11250688 PMCID: PMC521209 DOI: 10.1186/bcr24] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/1999] [Accepted: 11/18/1999] [Indexed: 11/14/2022] Open
Abstract
Advances in gene transfer technology have greatly expanded the opportunities for developing immunotherapy strategies for breast carcinoma. Genetic immunotherapy approaches include the transfer of genes encoding cytokines and costimulatory molecules to modulate immune function, as well as genetic immunization strategies which rely on the delivery of cloned tumor antigens. Improved gene transfer vectors, coupled with a better understanding of the processes that are necessary to elicit an immune response and an expanding number of target breast tumor antigens, have led to renewed enthusiasm that effective immunotherapy may be achieved. It is likely that immunotherapeutic interventions will find their greatest clinical application as adjuvants to traditional first-line therapies, targeting micrometastatic disease and thereby reducing the risk of cancer recurrence.
Collapse
Affiliation(s)
- T V Strong
- Gene Therapy Cewnter, Division of Heamtology-Oncology, University of Alabama at Birmingham, Birmingham, AL 35294-3300, USA.
| |
Collapse
|
23
|
Tüting T, Steitz J, Brück J, Gambotto A, Steinbrink K, DeLeo AB, Robbins P, Knop J, Enk AH. Dendritic cell-based genetic immunization in mice with a recombinant adenovirus encoding murine TRP2 induces effective anti-melanoma immunity. J Gene Med 1999; 1:400-6. [PMID: 10753065 DOI: 10.1002/(sici)1521-2254(199911/12)1:6<400::aid-jgm68>3.0.co;2-d] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The induction of cellular immune responses to melanocyte-specific enzymes such as the tyrosinase family of proteins is the goal of various clinical studies for the immunotherapy of melanoma. Tyrosinase-related protein-2 (TRP2) is an attractive model antigen for preclinical studies in C57BL/6 mice because it is naturally expressed by the murine B16 melanoma and can be recognized by self-reactive cytolytic T lymphocytes (CTL). Here we describe efforts to develop genetic immunization with dendritic cells (DC) for the immunotherapy of melanoma in this clinically relevant system. METHODS Recombinant adenoviruses encoding green fluorescent protein (Ad-EGFP) and murine TRP2 (Ad-mTRP2) were constructed using Cre-loxP-mediated recombination. DC were generated in vitro from precursors in bone marrow and transduced with Ad-EGFP or Ad-mTRP2. Mice were immunized by direct injection of adenovirus or by injection of Ad-transduced DC. Induction of tumor immunity was assessed by intravenous challenge with B16 melanoma cells and enumeration of experimentally induced lung metastases. RESULTS Flowcytometric analysis of DC transduced with Ad-EGFP demonstrated endogenous fluorescence due to cytoplasmatic expression of EGFP in 30-60% of cells. Ad-EGFP-transduced DC simultaneously displayed the DC-specific marker NLDC145 and high levels of MHC and costimulatory molecules on their cell surface. Transduction of DC with Ad-mTRP2 resulted in strong intracellular expression of TRP2 which could be readily detected by immunostaining. Importantly, immunization of mice with cultured Ad-mTRP2-transduced DC completely prevented the development of lung metastases following an intravenous challenge with B16 melanoma cells. This striking protective effect was observed with both the intravenous and the subcutaneous route of DC immunization. In vivo depletion of T-cell subsets suggested that the protective effect of an immunization with Ad-mTRP2-transduced DC involved both CD8+ and CD4+ T-cells. CONCLUSIONS Our results demonstrate that DC-based genetic immunization of mice with TRP2, a clinically relevant melanocyte-specific self-antigen, induces effective cellular immunity and prevents metastatic growth of B16 melanoma cells in vivo.
Collapse
Affiliation(s)
- T Tüting
- Department of Dermatology, J. Gutenberg-University, Mainz, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|