Breaking Immune Tolerance to Granule Proteases with Full-Length Antigen Vaccine in Humanized Transgenic Mice Reveals Alternative Antigen Processing and Immunodominance Heirarchy Applicable to Clinical Immunotherapy.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2054-2054
Author(s):  
J. Diamond Don ◽  
Wendi Zhou ◽  
Tumul Srivastava ◽  
Ravindra Rawal ◽  
Katharine Hagan ◽  
...  

Abstract Abstract 2054 Poster Board II-31 The serine proteinases, human neutrophil elastase (HNE) and proteinase 3 (PR3) are degradative enzymes stored in cytoplasmic azurophilic granules of neutrophils. Both proteins are aberrantly expressed in human myeloid leukemias. Over-expression of PR3 is important in maintaining a leukemia phenotype, while HNE may suppress hematopoietic progenitors. PR3 and HNE contain a conserved nonameric HLA-A*02 restricted T-cell epitope called PR1. The presence of PR1-specific CTL has been correlated with molecular remission in CML. A PR1 nonameric peptide vaccine has been evaluated clinically, with promising results. A limitation of the PR1 peptide vaccine is its restriction to patients with the HLA-A*02 allele. A vaccine comprising a larger portion of the PR3 and/or HNE protein and capable of inducing broader T-cell responses restricted by a wider range of HLA alleles is a potential alternative. We have generated numerous recombinant vaccinia viruses (rVV) expressing forms of PR3 and HNE and have evaluated CTL responses in an HLA-A*02 transgenic mouse model (Tg-A2): (1) rVV-EGFP-PR3, contains a gene encoding EGFP fused to human PR3 lacking the N-terminal signal peptide, while retaining both pro-dipeptide and C-terminal propeptide sequences; (2) EGFP fused to C-terminal truncated PR3 (rVV-EGFP-PR3-T) deleted of PR2 CTL epitope, but maintaining PR1 and; (3) rVV-EGFP-HNE. Tg-A2 were immunized with rVV-EGFP-PR3, and 2 weeks later immune splenocytes were expanded by in vitrostimulation (IVS) with syngeneic naïve mouse splenocytes loaded with an overlapping PR3 peptide library. Intracellular cytokine (ICC) assays using a PR3 peptide library or PR1 9-mer epitope peptide demonstrated a robust PR3-specific CD8+ T-cell response, surprisingly without a detectable response to the PR1 peptide. However, PR2, a decamer, is immunodominant in mice following this immunization. This result was unexpected, since the PR2 epitope was identified by computer prediction as a potential HLA-A*02-restricted epitope within PR3 but, unlike the HLA-A*02-restricted 9-mer epitope PR1, has not been extensively studied as a T-cell epitope in humans. We tested whether the immunodominant PR2-specific response in rVV-EGFP-PR3 immunized mice is also found when a naturally-processed PR3 protein is used as the stimulating antigen, expressed from the cell line K562-A2-PR3, a derivative of K562-A2 endogenously expressing human PR3. Splenocytes from mice immunized with rVV-EGFP-PR3 were expanded by co-culture with irradiated K562-A2-PR3 cells, then tested in ICC assays using PR1 and PR2 CTL epitope peptides. We observed a robust response to PR2 but not PR1. While the PR1 epitope is absolutely conserved between human and murine versions of PR3 and HNE, the PR2 epitope is unique to human PR3, diverging from murine PR3 by 3 amino acids, and by 5 amino acid from human and murine HNE. We speculated that Tg-A2 mice may be tolerant to PR1 due to the presence of this epitope within murine PR3 and HNE, which may account for dominance of PR2-specific responses. To investigate the possibility of inducing PR1-specific CTL in Tg-A2, mice were immunized with rVV-EGFP-HNE, and we detected PR1-specific responses by IVS followed by ICC with the PR1 peptide. These results indicate that tolerance to PR1 can likely be broken by immunization with rVV expressing HNE protein that contains the PR1 epitope. These observations can be explained by either the PR2>PR1 competition hypothesis, or by different amino acid sequences flanking the PR1 epitope in PR3 and HNE proteins affecting processing of the protein to generate the PR1 peptide. To test this question, we used rVV-PR3-T to immunize Tg-A2 mice, then conducted IVS followed by ICC with the PR1 peptide to detect PR1-specific CTL. Results showed that immunization with rVV expressing truncated PR3 reliably induced PR1-specific CTL, unlike immunization with rVV expressing the full-length PR3 antigen. We concluded that PR2 epitope prevented the recognition of PR1. Therefore in Tg-A2 mice, vaccination with poxvirus vaccines expressing PR3 will induce an HLA-A2-restricted CTL response almost entirely focused on PR2, whereas immunization with rVV-HNE will induce PR1-specific CTL. If this were replicated in humans, the majority of PR1-specific CTL detected in CML patients could be derived from HNE rather than PR3 antigen presentation, and that PR2-specific CTL derived from PR3 should be equally frequent as PR1 and likely contribute to clinical GVL and tumor regression. Disclosures: No relevant conflicts of interest to declare.

1991 ◽  
Vol 174 (2) ◽  
pp. 425-434 ◽  
Author(s):  
K Falk ◽  
O Rötzschke ◽  
K Deres ◽  
J Metzger ◽  
G Jung ◽  
...  

Virus-specific cytotoxic T lymphocytes (CTL) recognize virus-derived peptides presented by major histocompatibility complex (MHC) class I molecules on virus-infected cells. Such peptides have been isolated from infected cells and were compared to synthetic peptides. We found previously the Kd- or Db-restricted natural influenza nucleoprotein peptides to coelute on reversed phase high performance liquid chromatography columns with certain peptidic by-products present in synthetic peptide preparations. Here we show by extensive biochemical and immunological comparison that the natural peptides in all respects behave as the surmised synthetic nonapeptides, and thus, must be identical to them. The absolute amounts of these natural peptides contained in infected cells could be determined to be between 220 and 540 copies by comparing with defined amounts of pure synthetic nonapeptides. The comparison of the natural Kd-restricted peptide with published synthetic peptides known to contain other Kd-restricted CTL epitopes suggested a new MHC allele-specific T cell epitope forecast method, based on the defined length of nine amino acid residues and on critical amino acid residues at the second and the last position.


1998 ◽  
Vol 72 (8) ◽  
pp. 6965-6965
Author(s):  
Byung S. Kim ◽  
Robert L. Yauch ◽  
Young Yil Bahk ◽  
Jeong-Ah Kang ◽  
Mauro C. Dal Canto ◽  
...  

2020 ◽  
Vol 21 (4) ◽  
pp. 325-340 ◽  
Author(s):  
Saeed Anwar ◽  
Jarin T. Mourosi ◽  
Md. Fahim Khan ◽  
Mohammad J. Hosen

Background: Chikungunya is an arthropod-borne viral disease characterized by abrupt onset of fever frequently accompanied by joint pain, which has been identified in over 60 countries in Africa, the Americas, Asia, and Europe. Methods: Regardless of the availability of molecular knowledge of this virus, no definite vaccine or other remedial agents have been developed yet. In the present study, a combination of B-cell and T-cell epitope predictions, followed by molecular docking simulation approach has been carried out to design a potential epitope-based peptide vaccine, which can trigger a critical immune response against the viral infections. Results: A total of 52 sequences of E1 glycoprotein from the previously reported isolates of Chikungunya outbreaks were retrieved and examined through in silico methods to identify a potential B-cell and T-cell epitope. From the two separate epitope prediction servers, five potential B-cell epitopes were selected, among them “NTQLSEAHVEKS” was found highly conserved across strains and manifests high antigenicity with surface accessibility, flexibility, and hydrophilicity. Similarly, two highly conserved, non-allergenic, non-cytotoxic putative T-cell epitopes having maximum population coverage were screened to bind with the HLA-C 12*03 molecule. Molecular docking simulation revealed potential T-cell based epitope “KTEFASAYR” as a vaccine candidate for this virus. Conclusion: A combination of these B-cell and T-cell epitope-based vaccine can open up a new skyline with broader therapeutic application against Chikungunya virus with further experimental and clinical investigation.


2000 ◽  
Vol 68 (6) ◽  
pp. 3385-3393 ◽  
Author(s):  
Hiroko Asahi ◽  
Ahmed Osman ◽  
Rosemary M. Cook ◽  
Philip T. LoVerde ◽  
Miguel J. Stadecker

ABSTRACT In schistosomiasis mansoni, hepatic granulomatous inflammation surrounding parasite eggs is mediated by CD4+ T helper (Th) cells sensitized to schistosomal egg antigens (SEA). We previously showed that a prominent lymphoproliferative response of CD4+ Th cells from schistosome-infected C57BL/6 (BL/6) mice was directed against a 62-kDa component of SEA. A partial amino acid sequence of the 62-kDa component was found to be identical with one present in the enzyme phosphoenolpyruvate carboxykinase (PEPCK). Based on this sequence, a cDNA clone containing the entire coding region of PEPCK was identified, and the full recombinant Schistosoma mansoni PEPCK (rSm-PEPCK) of 626 amino acids was purified from a prokaryotic expression system. rSm-PEPCK strongly stimulated a specific T-cell hybridoma, 4E6, as well as CD4+ Th cells from SEA-immunized BL/6 mice and from infected BL/6, CBA, and BALB/c mice. In the infected mice, rSm-PEPCK elicited significant gamma interferon production as well as, to a lesser extent, production of interleukin-2 and -5. In BL/6 and BALB/c mice, the CD4+ Th cell response to rSm-PEPCK was greater than that directed against the egg antigen Sm-p40; conversely, CBA mice responded better to Sm-p40 than to Sm-PEPCK. A 12-amino-acid region (residues 398 to 409: DKSKDPKAHPNS) was demonstrated to contain a T-cell epitope; synthetic peptides containing this epitope significantly stimulated specific hybridoma 4E6 and polyclonal CD4+ Th cells. The identification and characterization of immunogenic egg components will contribute to the understanding and possible control of T-cell-mediated schistosomal disease.


2019 ◽  
Vol 1 (12) ◽  
pp. 760-772 ◽  
Author(s):  
Akiko Tenma ◽  
Hironori Nakagami ◽  
Hideki Tomioka ◽  
Makoto Sakaguchi ◽  
Ryoko Ide ◽  
...  

2001 ◽  
Vol 276 (27) ◽  
pp. 24525-24530 ◽  
Author(s):  
Stefan Reinelt ◽  
Merce Marti ◽  
Séverine Dédier ◽  
Thomas Reitinger ◽  
Gerd Folkers ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document