首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
We have demonstrated that coupling an immunoregulatory segment of the MHC class II-associated invariant chain (Ii), the Ii-Key peptide, to a promiscuous MHC class II epitope significantly enhances its presentation to CD4+ T cells. Here, a series of homologous Ii-Key/HER-2/neu(776-790) hybrid peptides, varying systematically in the length of the epitope(s)-containing segment, are significantly more potent than the native peptide in assays using T cells from patients with various types of tumors overexpressing HER-2/neu. In particular, priming normal donor and patient PBMCs with Ii-Key hybrid peptides enhances recognition of the native peptide either pulsed onto autologous dendritic cells (DCs) or naturally presented by IFN-gamma-treated autologous tumor cells. Moreover, patient-derived CD4+ T cells primed with the hybrid peptides provide a significantly stronger helper effect to autologous CD8+ T cells specific for the HER-2/neu(435-443) CTL epitope, as illustrated by either IFN-gamma ELISPOT assays or specific autologous tumor cell lysis. Hybrid peptide-specific CD4+ T cells strongly enhanced the antitumor efficacy of HER-2/neu(435-443) peptide-specific CTL in the therapy of xenografted SCID mice inoculated with HER-2/neu overexpressing human tumor cell lines. Our data indicate that the promiscuously presented vaccine peptide HER-2/neu(776-790) is amenable to Ii-Key-enhancing effects and supports the therapeutic potential of vaccinating patients with HER-2/neu+ tumors with such Ii-Key/HER-2/neu(776-790) hybrid peptides.  相似文献   

2.
Our aim is to develop peptide vaccines that stimulate tumor antigen-specific T-lymphocyte responses against frequently detected cancers. We describe herein a novel HLA-A*0201-restricted epitope, encompassing amino acids 828–836 (residues QIAKGMSYL), which is naturally presented by various HER-2/neu + tumor cell lines. HER-2/neu(828-836), [HER-2(9828)], possesses two anchor residues and stabilized HLA-A*0201 on T2 cells in a concentration-dependent Class I binding assay. This peptide was stable for 3.5 h in an off-kinetic assay. HER-2(9828) was found to be immunogenic in HLA-A*0201 transgenic (HHD) mice inducing peptide-specific and functionally potent CTL and long-lasting anti-tumor immunity. Most important, using HLA-A*0201 pentamer analysis we could detect increased ex vivo frequencies of CD8+ T-lymphocytes specifically recognizing HER-2(9828) in 8 out of 20 HLA-A*0201+ HER-2/neu + breast cancer patients. Moreover, HER-2(9828)-specific human CTL recognized the tumor cell line SKOV3.A2 as well as the primary RS.A2.1.DR1 tumor cell line both expressing HER-2/neu and HLA-A*0201. Finally, therapeutic vaccination with HER-2(9828) in HHD mice was proven effective against established transplantable ALC.A2.1.HER tumors, inducing complete tumor regression in 50% of mice. Our data encourage further exploitation of HER-2(9828) as a promising candidate for peptide-based cancer vaccines.  相似文献   

3.
We investigated the ubiquitination and degradation of a tumor antigen, the HER-2/neu (HER-2) protooncogene product which is overexpressed in epithelial cancers. HER-2 degradation was investigated in the ovarian tumor line, SKOV3.A2, that constitutively overexpressed long-life HER-2. We used as agonist geldanamycin (GA), which initiated downmodulation of HER-2 from the cell surface. HER-2 was polyubiquitinated and degraded faster in the presence than in the absence of GA. GA did not decrease HLA-A2 expression. Presentation of the immunodominant cytotoxic T lymphocyte (CTL) epitope, E75 (369–377) from SKOV.A2 was inhibited by proteasome inhibitors, such as LLnL but was enhanced by cysteine protease inhibitors such as E64, indicating that both the proteasome and cysteine proteases are involved in epitope formation but have different effects. Enhanced tumor recognition was not an immediate or early effect of GA treatment, but was evident after 20 h of GA treatment. In contrast, 20 h GA treatment did not increase tumor sensitivity to LAK cell lysis. Twenty hour GA-treated SKOV3.A2 cells expressed an unstable HER-2 protein synthesized in the presence of GA, of faster electrophoretic mobility than control HER-2. This suggested that the newly synthesized HER-2 in the presence of GA was the main source of epitopes recognized by CTL. Twenty hour GA-treated SKOV3.A2 cells were better inducers of CTL activity directed to a number of HER-2 CTL epitopes, in peripheral blood mononuclear cells compared with control untreated SKOV3.A2 cells. Thus, induction of HER-2 protein instability enhanced the sensitivity of tumor for CTL lysis. Increased HER-2 CTL epitopes presentation may have implications for overcoming the poor immuno-genicity of human tumors, and design of epitope precursors for cancer vaccination.  相似文献   

4.
To broaden the applicability of adoptive cellular immunotherapy against HER-2/neu overexpressing human cancers, we constructed a chimeric scFv/gamma gene composed of the variable regions of a HER-2/neu specific monoclonal antibody (mAb) joined to the signaling gamma-chain of the Fc(epsilon)RI receptor. The scFv(anti-HER-2/neu)/gamma chimeric gene was successfully expressed as functional surface receptor in the MD.45 cytolytic T-cell (CTL) hybridoma (MD.45-HER/gamma). Expression of the chimeric protein triggered IL-2 and IFN-gamma secretion in vitro upon encountering cell surface HER-2/neu and mediated non-major-histocompatibility-complex (MHC)-restricted HER-2/neu-specific target cell lysis. We also examined the in vivo activity of the MD.45-HER/gamma transduced cells. Severe combined immunodeficiency disease (SCID) mice that were given HER-2/neu positive (+) human tumor cell lines had significantly increased survival compared to mice treated with saline only, or with MD.45 cells transduced with a control anti-trinitrophenyl (anti-TNP) chimeric receptor gene (MD.45-TNP/gamma). These results demonstrate the feasibility of redirecting MD.45 CTL to react in vitro and in vivo with a variety of HER-2/neu(+) tumor cells by our gene transduction protocol. Moreover, they open the possibility of using the same chimeric gene for transducing primary lymphocytes and thus allowing adoptive immunotherapy against HER-2/neu(+) cancers.  相似文献   

5.
In order to broaden the possibility for anti-HER-2/neu (HER-2) immune targeting, it is important to identify HLA-A24 restricted peptide epitopes derived from HER-2, since HLA-A24 is one of the most common alleles in Japanese and Asian people. In the present study, we have screened HER-2-derived, HLA-A24 binding peptides for cytotoxic T lymphocyte (CTL) epitopes. A panel of HER-2-derived peptides with HLA-A24 binding motifs and the corresponding analogs designed to enhance HLA-A24 binding affinity were selected. Identification of HER-2-reactive and HLA-A24 restricted CTL epitopes were performed by a reverse immunology approach. To induce HER-2-reactive and HLA-A24 restricted CTLs, PBMCs from healthy donors were repeatedly stimulated with monocytes-derived, mature DCs pulsed with HER-2 peptide. Subsequent peptide-induced T cells were tested for the specificity by enzyme linked immunospot, cytotoxicity and tetramer assays. CTL clones were then obtained from the CTL lines by limiting dilution. Of the peptides containing HLA-A24 binding motifs, 16 peptides (nine mers) including wild type peptides (IC50<1,000 nM) and substituted analog peptides (IC50<50 nM) were selected for the present study. Our studies show that an analog peptide, HER-2(905AA), derived from HER-2(905) could efficiently induce HER-2-reactive and HLA-A24 restricted CTLs. The reactivity of the HER-2(905AA)-induced CTL (CTL905AA) was confirmed by different CTL assays. The CTL905AA clones also were able to lyse HER-2(+), HLA-A24(+) tumor cells and cytotoxicity could be significantly reduced in cold target inhibition assays using cold targets pulsed with the HER-2(905) wild type peptide as well as the inducing HER-2(905AA) analog peptide. A newly identified HER-2(905) peptide epitope is naturally processed and presented as a CTL epitope on HER-2 overexpressing tumor cells, and an MHC anchor-substituted analog, HER-2(905AA), can efficiently induce HER-2-specific, HLA-A24 restricted CTLs.  相似文献   

6.
HER-2/neu oncoprotein is overexpressed in a variety of human tumors and is associated with aggressive disease. Immunogenic HER-2/neu CTL epitopes have been used as vaccines for the treatment of HER-2/neu positive malignancies with limited success. By applying prediction algorithms for MHC class I ligands and proteosomal cleavages, in this study, we describe the identification of HER-2/neu decamer LIAHNQVRQV spanning residues 85-94 (HER-2(10(85))). HER-2(10(85)) proved to bind with high affinity to HLA-A2.1 and was stable for 4 h in an off-kinetics assay. This peptide was immunogenic in HLA-A2.1 transgenic (HHD) mice inducing peptide-specific CTL, which responded to tumor cell lines of various origin coexpressing human HER-2/neu and HLA-A2.1. This demonstrates that HER-2(10(85)) is naturally processed from endogenous HER-2/neu. Five of sixteen HER-2/neu+ HLA-A2.1+ breast cancer patients analyzed had HER-2(10(85))-reactive T cells ranging from 0.35-0.70% of CD8+ T cells. Depletion of T regulatory cells from PBMC enabled the rapid expansion of HLA-A2.1/HER-2(10(85))pentamer+/CD8+ cells (PENT+/CD8+), whereas significantly lower numbers of CTL could be generated from unfractionated PBMC. HER-2(10(85))-specific human CTL recognized the HER-2/neu+ HLA-A2.1+ tumor cell line SKBR3.A2, as determined by IFN-gamma intracellular staining and in the high sensitivity CD107alpha degranulation assay. Finally, HER-2(10(85)) significantly prolonged the survival of HHD mice inoculated with the transplantable ALC.A2.1.HER tumor both in prophylactic and therapeutic settings. These data demonstrate that HER-2(10(85)) is an immunogenic peptide, capable of eliciting CD8-mediated responses in vitro and in vivo, providing the platform for further exploitation of HER-2(10(85)) as a possible target for anticancer immunotherapy.  相似文献   

7.
HER-2/neu oncoprotein is overexpressed in a variety of human tumors and is associated with malignant transformation and aggressive disease. Due to its overexpression in tumor cells and because it has been shown to be immunogenic, this protein represents an excellent target for T-cell immunotherapy. Peptide extracts derived from primary HLA-A*0201-positive (+) HER-2/neu+ human tumors by acid elution (acid cell extracts (ACEs)) were tested for their capacity to elicit in HLA-A*0201 transgenic mice, cytotoxic T lymphocytes (CTLs) lysing HLA-A*0201+ HER-2/neu+ tumor cells. Injections of ACE in transgenic mice induced CTLs capable of specifically lysing HER-2/neu+ tumor cell lines (also including the original HER-2/neu+ primary tumor cells from which the ACEs were derived) in an HLA-A*0201–restricted fashion. Adoptive transfer of ACE-induced CTLs was sufficient to significantly prolong survival of SCID mice inoculated with HLA-A*0201+ HER-2/neu+ human tumor cell lines. Cytotoxicity of such ACE-induced CTL lines was directed, at least as detected herein, also against the HER-2/neu peptides HER-2 (9369) and HER-2 (9435) demonstrating the immunodominance of these epitopes. HER-2 peptide–specific CTLs generated in the HLA-A*0201–transgenic mice, upon peptide immunization, lysed in vitro HER-2/neu+ human tumor cell lines in an HLA-A*0201–restricted manner and, when adoptively transferred, conferred sufficient protection in SCID mice inoculated with the same human tumor cell lines as above. However, CTLs induced by ACEs displayed enhanced efficacy in the therapy of xenografted SCID mice compared with the HER-2 peptide–specific CTLs (i.e., HER-2 [9369] or HER-2 [9435]). Even by administering mixtures of CTLs specific for each of these peptides, the prolongation of survival achieved was still inferior compared with that obtained with ACE-induced CTLs. This suggested that additional epitopes may contribute to the immunogenicity of such tumor-derived ACEs. Thus, immunization with ACEs from HER-2/neu+ primary tumor cells appears to be an effective approach to generate multiple and potent CTL-mediated immune responses against HER-2/neu+ tumors expressing the appropriate HLA allele(s). By screening ACE-induced CTL lines with synthetic peptides encompassing the HER-2/neu sequence, it is feasible to identify immunodominant epitopes which may be used in mixtures as vaccines with enhanced efficacy in both the prevention and therapy of HER-2/neu+ malignancies.This work was supported by grants from the Regional Operational Program Attika (No. 20, MIS code 59605GR) to M.P., and from the GSRT Program (No. PENED 01ED55) to C.N.B.  相似文献   

8.
Simian virus 40 large tumor (T) antigen contains three H-2Db-restricted (I, II/III, and V) and one H-2Kb-restricted (IV) cytotoxic T lymphocyte (CTL) epitopes. We demonstrate that a hierarchy exists among these CTL epitopes, since vigorous CTL responses against epitopes I, II/III, and IV are detected following immunization of H-2b mice with syngeneic, T-antigen-expressing cells. By contrast, a weak CTL response against the H-2Db-restricted epitope V was detected only following immunization of H-2b mice with epitope loss variant B6/K-3,1,4 cells, which have lost expression of CTL epitopes I, II/III, and IV. Limiting-dilution analysis confirmed that the lack of epitope V-specific CTL activity in bulk culture splenocytes correlated with inefficient expansion and priming of epitope V-specific CTL precursors in vivo. We examined whether defined genetic alterations of T antigen might improve processing and presentation of epitope V to the epitope V-specific CTL clone Y-5 in vitro and/or overcome the recessive nature of epitope V in vivo. Deletion of the H-2Db-restricted epitopes I and II/III from T antigen did not increase target cell lysis by epitope V-specific CTL clones in vitro. The amino acid sequence SMIKNLEYM, which species an optimized H-2Db binding motif and was found to induce CTL in H-2b mice, did not further reduce epitope V presentation in vitro when inserted within T antigen. Epitope V-containing T-antigen derivatives which retained epitopes I and II/III or epitope IV did not induce epitope V-specific CTL in vivo: T-antigen derivatives in which epitope V replaced epitope I failed to induce epitope V-specific CTL. Recognition of epitope V-H-2Db complexes by multiple independently derived epitope V-specific CTL clones was rapidly and dramatically reduced by incubation of target cells in the presence of brefeldin A compared with the recognition of the other T-antigen CTL epitopes by epitope specific CTL, suggesting that the epitope V-H-2Db complexes either are labile or are present at the cell surface at reduced levels. Our results suggest that processing and presentation of epitope V is not dramatically altered (reduced) by the presence of immunodominant CTL epitopes in T antigen and that the immunorecessive nature of epitope V is not determined by amino acids which flank its native location within simian virus 40 T antigen.  相似文献   

9.
The regulatory immediate-early (IE) protein pp89 of murine cytomegalovirus induces CD8+ T lymphocytes that protect against lethal murine cytomegalovirus infection. The IE1 epitope is the only epitope of pp89 that is recognized by BALB/c cytolytic T lymphocytes (CTL). Using synthetic peptides, the optimal and minimal antigenic sequences of the IE1 epitope have been defined. To evaluate the predictive value of data obtained with synthetic peptides, recombinant vaccines encoding this single T-cell epitope were constructed using as a vector the hepatitis B virus core antigen encoded in recombinant vaccinia virus. In infected cells expressing the chimeric proteins, only IE1 epitope sequences that were recognized as synthetic peptides at concentrations lower than 10(-6) M were presented to CTL. Vaccination of mice with the recombinant vaccinia virus that encoded a chimeric protein carrying the optimal 9-amino-acid IE1 epitope sequence elicited CD8+ T lymphocytes with antiviral activity and, furthermore, protected against lethal disease. The results thus show for the first time that recombinant vaccines containing a single foreign nonameric CTL epitope can induce T-lymphocyte-mediated protective immunity.  相似文献   

10.
Five overlapping fragments of rat HER-2/neu have been expressed in recombinant Listeria monocytogenes. Each fragment of HER-2/neu is secreted as a fusion protein with a truncated, nonhemolytic form of listeriolysin O (LLO). Lm-LLO-EC1, Lm-LLO-EC2, and Lm-LLO-EC3 overlap the extracellular domain of HER-2/neu, whereas Lm-LLO-IC1 and Lm-LLO-IC2 span the intracellular domain. All five strains controlled the growth of established NT-2 tumors, a rat HER-2/neu-expressing tumor line derived from a spontaneously arising mammary tumor in a FVB/N HER-2/neu-transgenic mouse. The antitumor effect of each of these vaccine constructs was abrogated by the in vivo depletion of CD8(+) T cells, although only one known epitope has been defined previously and is present in Lm-LLO-EC2. Anti-HER-2/neu CTL responses were generated by each of the rLm vaccine constructs. With the use of a panel of 3T3 cell lines expressing overlapping fragments of HER-2/neu, regions of HER-2/neu with potential CD8(+) T cell epitopes have been defined. DNA vaccines expressing either a fragment or full-length HER-2/neu were constructed in LLO-fused and non-LLO-fused forms. CTL analysis of the DNA vaccines revealed a broadening in the regions of HER-2/neu recognizable as targets when the target Ag is fused to LLO. These studies show the efficacy of L. monocytogenes-based HER-2/neu vaccines in a murine model of breast cancer and also that the immunogenicity of self-Ags can be increased by fusion to LLO and delivery by L. monocytogenes revealing subdominant epitopes.  相似文献   

11.
CTL are important in combating cancer and viruses. Therefore, triggering the complete potential of CTL effector functions by new vaccination strategies will not only improve prophylaxis of tumor or virus-related diseases, but also open opportunities for effective therapeutic immunizations. Using transcutaneous immunization, we show that epicutaneous (e.c.)(4) application of an ointment containing a CTL epitope and the TLR7 ligand imiquimod is highly effective in activating T cells in mice using TCR-transgenic CTL or in wild-type mice. Transcutaneous immunization-activated CTL mount a full-blown immune response against the target epitope characterized by proliferation, cytolytic activity, and the production of IFN-gamma that is completely restricted to the epitope used for vaccination. Our results obtained by simple e.c. application of an ointment, without further skin irritating procedures, provide the basis for the development of new, easy to use vaccines against cancer or virus-associated diseases.  相似文献   

12.
The HER-2/neu (HER-2) oncogene is expressed in normal epithelial surfaces at low levels and overexpressed in several types of tumors. The low immunogenicity against this self tumor Ag can be improved by developing epitopes with amino acid replacements in their sequences. In this study, three HER-2/neu.369 (HER-2.369) analogue peptides, produced by modifying both anchor positions by introducing L, V, or T at position 2 and V at the C terminus, were analyzed for their capacity to induce CTLs in vitro from human PBMC and in vivo in HLA-A2.1/Kb transgenic mice. One of the analogues (HER-2.369 V2V9) sensitized target cells for HER-2-specific recognition by human CTLs and induced specific CTLs in vitro at 100-fold lower concentrations than the HER-2.369 wild-type epitope. These CTLs were also able to recognize the wild-type epitope and HER-2-expressing tumors in an MHC-restricted manner. Furthermore, a 100-fold lower amount of the HER-2.369 V2V9 analogue compared with the wild-type epitope was required to induce CTLs in HLA-A2.1/Kb transgenic mice. However, the V2V9 analogue demonstrated only marginally better binding to the MHC class I A2 allele compared with wild type. To establish thermodynamic parameters, we developed radiolabeled F3*Y analogues from both the HER-2.369 epitope and the V2V9 analogue. Our results indicate that the high biological activity of the HER-2.369 V2V9 epitope is associated with a slower dissociation kinetic profile, resulting in an epitope with greater HLA-A2 stability.  相似文献   

13.
Liu W  Zhai M  Wu Z  Qi Y  Wu Y  Dai C  Sun M  Li L  Gao Y 《Amino acids》2012,42(6):2257-2265
Identification of cytotoxic T lymphocyte (CTL) epitopes from tumor antigens is essential for the development of peptide vaccines against tumor immunotherapy. Among all the tumor antigens, the caner-testis (CT) antigens are the most widely studied and promising targets. PLAC1 (placenta-specific 1, CT92) was considered as a novel member of caner-testis antigen, which expressed in a wide range of human malignancies, most frequently in breast cancer. In this study, three native peptides and their analogues derived from PLAC1 were predicted by T cell epitope prediction programs including SYFPEITHI, BIMAS and NetCTL 1.2. Binding affinity and stability assays in T2 cells showed that two native peptides, p28 and p31, and their analogues (p28-1Y9?V, p31-1Y2L) had more potent binding activity towards HLA-A*0201 molecule. In ELISPOT assay, the CTLs induced by these four peptides could release IFN-γ. The CTLs induced by these four peptides from the peripheral blood mononuclear cells (PBMCs) of HLA-A*02+ healthy donor could lyse MCF-7 breast cancer cells (HLA-A*0201+, PLAC1+) in vitro. When immunized in HLA-A2.1/Kb transgenic mice, the peptide p28 could induce the most potent peptide-specific CTLs among these peptides. Therefore, our results indicated that the peptide p28 (VLCSIDWFM) could serve as a novel candidate epitope for the development of peptide vaccines against PLAC1-positive breast cancer.  相似文献   

14.
HER-2/neu is an immunogenic protein eliciting both humoral and cellular immune responses in patients with HER-2/neu-positive (+) tumors. Preexisting cytotoxic T lymphocyte (CTL) immunity to HER-2/neu has so far been mainly evaluated in terms of detection of CTL precursor (CTLp) frequencies to the immunogenic HLA-A2–binding nona-peptide 369-377 (HER-2(9369)). In the present study, we examined patients with HER-2/neu+ breast, ovarian, lung, colorectal, and prostate cancers for preexisting CTL immunity to four recently described HER-2/neu–derived and HLA-A2–restricted "cytotoxic" peptides and to a novel one spanning amino acids 777–785 also with HLA-A2–binding motif. We utilized enzyme-linked immunosorbent spot (ELISpot) assay, which allows a quantitative and functional assessment of T cells directed against specific peptides after only brief in vitro incubation. CTL reactivity was determined with an interferon (IFN-) ELISpot assay detecting T cells at the single cell level secreting IFN-. CTLp were defined as peptide-specific precursors per 106 peripheral blood mononuclear cells (PBMCs). Patients' PBMCs with increased CTLp were also tested against autologous tumor targets and peptide-pulsed dendritic cells (DCs) in cytotoxicity assays. We also studied patients with HER-2/neu-negative (-) tumors and healthy individuals. Of the HER-2/neu+ patients examined, 31% had increased CTLp to HER-2(9952), 19% to HER-2(9665), 16% to HER-2(9689), and 12.5% HER-2(9435), whereas only 2 of 32 patients (6%) responded to HER-2(9777). The CTLp recognizing HER-2(9952) were extremely high in two patients with breast cancer, one with lung cancer, and one with prostate cancer. None of the HER-2/neu- patients or healthy donors exhibited increased CTLp to any of these peptides. Besides IFN- production, preexisting CTL immunity to all five HER-2/neu peptides was also shown in cytotoxicity assays where patients' PBMCs with increased CTLp specifically lysed autologous tumor targets and autologous peptide-pulsed DCs. Our results demonstrate for the first time that (1) preexisting immunity to peptides HER-2(9435), HER-2(9952), HER-2(9689), HER-2(9665), and HER-2(9777) is present in patients with HER-2/neu+ tumors of distinct histology, (2) HER-2(9777) is a naturally processed peptide expressed on the surface of HER-2/neu+ tumors, as are the other four peptides, and (3) HER-2/neu+ prostate tumor cells can be recognized and lysed by autologous HER-2 peptide-specific CTL. Our findings broaden the potential application of HER-2/neu-based immunotherapy.  相似文献   

15.
The epitope corresponding to amino acid residues 147-161 of the nucleoprotein (NP) of influenza A virus is recognized by CTL in association with H-2Kd class I Ag. Herein, we engineered an Ig molecule carrying this CTL epitope by replacing the diversity gene segment of the H chain V region of an anti-arsonate antibody with an oligonucleotide that encodes the CTL epitope. The chimeric H chain gene was expressed either alone or together with the parental L chain in the nonsecreting BALB/c myeloma B cell line, SP2/0. The Ig produced by cells transfected with both the chimeric H chain and parental L chains genes expressed the NP epitope but lost the original arsonate binding activity. In addition, SP2/0 cells expressing the chimeric H chain either alone or together with the parental L chain were lysed by class I restricted NP-epitope specific CTL. By contrast, SP2/0 cells pulsed with soluble chimeric Ig molecules were not lysed by the specific CTL. These observations indicate that: 1) this particular CTL epitope can be expressed on Ig molecules without altering the H and L chain pairing; 2) this CTL epitope can be generated from this chimeric Ig in which it is surrounded by flanking regions distinct from those of the viral NP; and 3) the generation of this CTL epitope from the Ig molecule requires the endogenous pathway as do viral proteins.  相似文献   

16.
Mice which coexpress human papillomavirus type 16 E7 and HLA A2.1 in peripheral squamous epithelium and thymic cortical epithelium are tolerant at the cytotoxic T-lymphocyte (CTL) level to E7 epitopes restricted through HLA A*0201 and H-2b (T. Doan, M. Chambers, M. Street, G. J. Fernando, K. Herd, P. Lambert, and R. Tindle, Virology 244:352–364, 1998). Here we used bone marrow-reconstituted radiation chimeras to distinguish whether E7-directed CTL tolerance was mediated peripherally by E7 expression in skin or centrally by E7 expression in thymus. In chimeric mice expressing E7 in skin and reconstituted with E7-naïve bone marrow and E7-naïve thymus, CTL responses to vaccine-administered E7 epitopes were not restored, i.e., the mice remained tolerant. In contrast, chimeric mice not expressing E7 in skin and reconstituted with E7-naïve bone marrow and E7-expressing thymus had full E7-directed CTL responses. These results demonstrate that E7 protein expression in peripheral squamous epithelium is sufficient to tolerize the E7-directed CTL precursor repertoire. The data have implications for E7-mediated tumorigenesis and for the development of E7-based immunotherapeutic strategies, since peripheral immunological tolerance of tumor-associated antigens may create a barrier to effective immunotherapy.The E7 oncoprotein of human papillomavirus type 16 (HPV16) is a tumor-specific antigen when expressed in HPV16-associated cervical epithelial tumors, to which immunomanipulative strategies are being directed, both experimentally (see, for example, references 7, 9, and 29) and in E7-based therapeutic vaccine clinical trials (5). We recently reported studies with mice expressing HPV16 E7 protein, driven from the keratin 14 (K14) promoter, in basal epithelium of skin and in the thymic cortex (8). We showed that immunization-induced cytotoxic T-lymphocyte (CTL) responses to each of three CTL epitopes in the E7 protein restricted through two major histocompatibility complex (MHC) class 1 haplotypes were down-regulated in these E7-transgenic mice compared with non-E7 syngeneic control mice. However, in these studies we did not determine whether the down-regulation (i.e., tolerance) was induced centrally by E7 expressed in the thymus or peripherally by E7 expressed in epithelium. In the present study, we distinguish between these two possibilities by specific immunization of bone marrow-reconstituted thymus-transplanted chimeric E7 transgenic mice. We report that chimeric mice expressing the E7 transgene in peripheral epithelium but not in the thymus showed E7-specific down-regulated CTL responses to each of two E7 CTL epitopes restricted through a human and a mouse MHC class I allele, respectively, when compared with sham chimeric but non-E7 control mice. In contrast, chimeric mice expressing the E7 transgene in thymus, but not peripheral epithelium, showed E7-directed CTL responses indistinguishable from those of non-E7 control mice. Thus, we show that the expression of E7 in peripheral squamous epithelium is sufficient to induce and maintain a state of tolerance against E7.

E7-directed bone marrow-derived precursor CTLs (pCTLs) are not tolerized in mice expressing E7 in thymus but not in skin.

(K14E7 × A2.1Kb)F1 mice (designated KA mice) were derived by crossing male K14.HPV16E7(+/+) mice (16), which express an HPV16 E7 transgene perinatally and throughout life in skin and thymic cortical epithelium, with female HLA A2.1Kb(+/+) mice (30). (FVB × A2.1Kb)F1 mice (designated FA) are syngeneic but do not possess the E7 transgene. KA (E7+) and FA (E7) mice are on an H-2b background. To inquire whether pCTLs from E7-transgenic mice were tolerized on E7-expressing thymus, we constructed thymus-transplanted radiation chimeras as described elsewhere (8) from immunologically depleted FA (E7) mice reconstituted with KA (E7+) bone marrow cells. In half of the mice, [designated KA→FA(FA) mice], the bone marrow-derived T-cell precursors were made to mature through a thymus implant from an FA (E7) donor mouse; in the other half of the mice [designated KA→FA(KA) mice], the bone marrow cells were made to mature through an E7-expressing KA thymus implant (Fig. (Fig.1,1, panel I). KA→FA(FA) mice, KA→FA(KA) mice, and control FA (E7) and KA (E7+) mice were immunized for CTL response induction with a mix of peptides containing 82LLMGTLGIV90 (an HLA A*0201-restricted E7 CTL epitope [24]), 49RAHYNIVTF57 (an H-2Db-restricted E7 CTL epitope [9]), and 58GILGFVFTL66 (an HLA A*0201-restricted influenza virus matrix CTL epitope [13]). Control mice underwent surgical procedures but without receiving cell and/or organ transplants (sham). KA (E7+) mice showed the previously documented (8) down-regulated CTL response to the E7 epitopes (but not to the irrelevant influenza virus matrix epitope) compared to FA (E7) mice (Fig. (Fig.1,1, panels IIC and IID). In contrast, KA→FA(KA) mice exhibited E7 (and influenza virus matrix)-directed CTL responses of the same magnitude as those of KA→FA(FA) mice and FA (E7) mice (Fig. (Fig.1,1, panels IIA to IIC). These data indicate that E7-directed pCTLs from E7 transgenic mice which mature through an E7-expressing thymus, and emerge into a non-E7-expressing peripheral epithelial environment, are not tolerized. Open in a separate windowFIG. 1(I) Derivation of KA→FA(FA) and KA→FA(KA) chimeric mice from immunologically ablated FA (E7) mice. (II) CTL responses of splenocytes from chimeric mice and sham control FA (E7) and KA (E7+) mice (three per group) immunized with a mix of peptides containing E7 CTL epitopes LLMGTLGIV and RAHYNIVTF and influenza virus matrix CTL epitope GILGFVFTL. Immunizations were given in Quil A adjuvant and tetanus toxoid as described elsewhere (8). Spleen cells were restimulated with individual peptides in vitro. Targets were EL4.A2 cells (8) pulsed with individual peptides as indicated. EL4.A2 cells are susceptible to specific CTL lysis through both HLA A*0201 and H-2b restriction elements. CTL assays were conducted as described elsewhere (8). bm, bone marrow; th, thymus; sk, skin.To eliminate the possibility that bone marrow-derived precursors from KA (E7+) mice had somehow previously encountered E7 protein before transfer to recipient mice, thereby influencing their immunological status in the above-described experiment, we asked whether bone marrow-derived pCTLs from FA (E7) mice would be tolerized during maturation in an E7-expressing thymus. We constructed chimeras from immunologically ablated FA (E7) mice by reconstitution with FA (E7) bone marrow cells. In half the mice [designated FA→FA(KA) mice], the bone marrow cells were made to mature through an E7-expressing KA thymus implant. In the other half of the mice [designated FA→FA(FA) mice], the bone marrow cells were made to mature through a non-E7-expressing FA thymus implant (Fig. (Fig.2,2, panel I). FA→FA(KA) mice, FA→FA(FA) mice, and control FA (E7) and KA (E7+) mice were immunized for CTL induction with a mix of peptides containing the HLA A*0201-restricted and H-2b-restricted E7 CTL epitopes and influenza virus matrix CTL epitope. FA→FA(KA) mice exhibited E7-directed CTL responses of the same magnitude as those of FA→FA(FA) mice and FA (E7) mice, while KA (E7+) mice exhibited the expected down-regulated E7-directed (but not down-regulated influenza virus matrix-directed) CTL responses (Fig. (Fig.2,2, panel II). These data indicated that E7-naïve bone marrow-derived pCTLs which mature through an E7-expressing thymus and emerge into a non-E7 peripheral epithelial environment are not tolerized to E7. Open in a separate windowFIG. 2(I) Derivation of FA→FA(FA) and FA→FA(KA) chimeric mice from immunologically ablated FA (E7) mice. (II) CTL responses of splenocytes from chimeric mice and sham control mice (three per group) immunized with a mix of peptides containing E7 CTL epitopes LLMGTLGIV and RAHYNIVTF and influenza virus matrix CTL epitope GILGFVFTL. Spleen cells were restimulated with individual peptides in vitro. Targets were EL4.A2 cells pulsed with individual peptides as indicated. CTL assays were conducted as described elsewhere (8). bm, bone marrow; th, thymus; sk, skin.

Bone marrow-derived pCTLs are specifically tolerized in mice expressing E7 in skin but not in thymus.

To inquire whether E7-directed pCTLs were tolerized in mice expressing E7 in skin but not in thymus, we constructed chimeric mice in which bone marrow-derived precursors were made to mature through a non-E7-expressing thymus and to emerge into an E7-expressing peripheral epithelial environment. In a first experiment, immunologically ablated KA (E7+) mice were reconstituted with KA (E7+) bone marrow cells which were made to mature through a thymus implanted from an FA (E7) mouse. The recipient mice, designated KA→KA(FA) (Fig. (Fig.3,3, panel I), and control FA (E7) and KA (E7+) mice were immunized for CTL response induction with a mix of peptides containing the HLA A*0201-restricted and H-2b-restricted E7 CTL epitopes and the influenza virus matrix CTL epitope. In KA→KA(FA) mice, E7-directed CTL responses to both E7 epitopes were down-regulated to the level seen in control KA (E7+) mice (Fig. (Fig.3,3, panel IIB), while control FA (E7) mice showed the expected high responses to both E7 CTL epitopes. Open in a separate windowFIG. 3(I) Derivation of KA→KA(FA) chimeric mice from immunologically ablated KA (E7+) mice. (II) CTL responses of splenocytes from chimeric mice and sham control mice (three per group) immunized with a mix of peptides containing E7 CTL epitopes LLMGTLGIV and RAHYNIVTF and influenza virus matrix CTL epitope GILGFVFTL. Spleen cells were restimulated with individual peptides in vitro. Targets were EL4.A2 cells pulsed with individual peptides as indicated. CTL assays were conducted as described elsewhere (8). bm, bone marrow; th, thymus; sk, skin.In a second experiment, immunologically ablated KA (E7+) mice were reconstituted with bone marrow from FA (E7) mice, which was made to mature through a non-E7-expressing FA thymus. These mice, designated FA→KA(FA) mice (Fig. (Fig.4,4, panel I), were immunized for CTL response induction with a mix of peptides containing the HLA A*0201-restricted and the H-2b-restricted E7 CTL epitopes and the influenza virus matrix CTL epitope. As with KA→KA(FA) mice in the previous experiment, E7-directed CTL responses to both E7 epitopes were down-regulated as in KA (E7+) controls and in contrast to FA (E7) controls, while the influenza virus matrix response confirmed adequate reconstitution. Open in a separate windowFIG. 4(I) Derivation of FA→KA(FA) chimeric mice from immunologically ablated KA (E7+) mice. (II) CTL responses of splenocytes from chimeric mice and sham control mice (three per group) immunized once with a mix of peptides containing E7 CTL epitopes LLMGTLGIV and RAHYNIVTF and influenza virus matrix CTL epitope GILGFVFTL. Spleen cells were restimulated with individual peptides in vitro. Targets were EL4.A2 cells pulsed with individual peptides as indicated. CTL assays were conducted as described elsewhere (8). bm, bone marrow; th, thymus; sk, skin.The results from these two experiments indicate that bone marrow-derived E7-directed pCTLs which mature through a non-E7-expressing thymus and emerge into an E7-expressing epithelial environment are specifically tolerized to E7.We have previously reported pCTL tolerance to epitopes of the HPV16 E7 oncoprotein in KA mice expressing a K14 promoter-driven E7 transgene perinatally and throughout life in the thymus and in basal and/or suprabasal cells of peripheral epithelium (8). In the present experiments, we demonstrate that the E7-directed pCTL repertoire is tolerized in mice expressing E7 in peripheral epithelium in the absence of thymic expression. Conversely, the repertoire is not tolerized in mice expressing E7 in the thymus, in the absence of E7 expression in peripheral epithelium. These data indicate that expression of E7 in peripheral epithelium, and not the thymus, is sufficient to induce and maintain a state of pCTL tolerance to E7. In the thymus, the K14 promoter directs transgene expression to the cortical epithelial compartment (19), which, in other mouse models, has been shown to contribute to the shaping of the T-cell repertoire by positive rather than negative selection (for example, see reference 20). Melero et al. (21) observed no functional down-regulation of the CTL responses induced by immunization with E7 peptide epitope RAHYNIVTF in H-2b mice expressing HPV16 E7 from a K14 promoter and concluded that the mice remain immunologically ignorant of this epitope. This result contrasts with ours. Together, they provide further examples of T-cell tolerance to peripheral antigens in some systems (see, for example, references 1, 2, and 23) and T-cell ignorance in others (see, for example, references 14, 17, and 25). While determinants of immunological outcome of peripheral antigen expression are clearly complex (22), the level of expression (as well as timing and site of expression) can determine whether an antigen induces tolerance or is ignored by naïve T cells. This consideration may explain the difference between the results of Melero et al. and ours. The effect of the level of E7 expression on peripheral tolerance induction is under investigation in our laboratory.Specific CTL tolerance has implications for E7-mediated tumorigenesis. Nascent E7-expressing tumor cells will escape surveillance where little or no positive priming of cognate pCTLs by endogenous E7 occurs. Additionally, specific CTL tolerance which inhibits the generation of an immunization-induced CTL response will detract from effective immunotherapy (26). We have previously reported that (K14.E7 × C57)F1 mice fail to control a challenge with an E7-expressing tumor following immunization with E7 CTL epitope RAHYNIVTF, whereas in immunized non-E7-transgenic control mice the tumors did not become established (12). Failure to control the tumor was correlated with a lack of an inducible RAHYNIVTF-directed CTL response in E7-transgenic mice, in contrast to non-E7-transgenic control mice, where a powerful CTL response was observed. In further experiments, multiple immunization of KA mice with E7 CTL epitopes or whole E7 protein failed to arrest the development of E7-associated endogenous tumors (8), again being correlated with a lack of E7-directed CTL responses.The current therapeutic vaccine strategy for HPV16-associated cervical carcinoma targets the E7 tumor-specific antigen by CTL induction (5, 28). The possibility arises that chronic expression of E7 in transformed cervical epithelial cells during the life of the tumor functionally tolerizes E7-directed pCTLs.Ongoing experiments in our laboratory will distinguish between presentation of E7 to pCTLs directly by keratinocytes and cross presentation of exogenously acquired E7 by bone marrow-derived professional antigen-presenting cells. Presentation of antigen by either of these routes can be tolerogenic (3, 6, 15, 27). Additionally, we will determine whether loss of functional E7-directed CTLs results from pCTL deletion (4, 11) or anergy (10, 25). Peripheral tolerance of tissue-specific antigen depends, at least in some cases, on the generation of regulatory CD4+ cells (see, for example, reference 18). That E7-directed CTL tolerance in KA (E7+) mice reflects an impairment of cognate CD4+ help is unlikely in view of our finding that (K14.E7 × C57)F1 mice immunized with full-length E7 and displaying E7-specific pCTL tolerance showed concomitant enhanced E7-directed CD4+ T-helper responses (12).The data reported in the present study demonstrate the induction of peripheral tolerance in E7-directed pCTLs by HPV16 E7 expressed in squamous epithelial cells, in the context of human (and mouse) MHC class 1 haplotypes. There are direct implications for the development and progression of cervical cancers which express E7 in transformed squamous epithelium and for the design of E7-based immunotherapeutic strategies for cervical cancer. In the broader context, there are implications for CTL response induction to any foreign or aberrant protein expressed constitutively in squamous epithelial cells as a result of infection, tumorigenesis, or appearance of autoantigen.  相似文献   

17.
Trastuzumab is a growth-inhibitory humanized Ab targeting the oncogenic protein HER-2/neu. Although trastuzumab is approved for treatment of advanced breast cancer, a number of concerns exist with passive immunotherapy. Treatment is expensive and has a limited duration of action, necessitating repeated administrations of the mAb. Active immunotherapy with conformational B cell epitopes affords the possibility of generating an enduring immune response, eliciting protein-reactive high-affinity anti-peptide Abs. The three-dimensional structure of human HER-2 in complex with trastuzumab reveals that the Ag-binding region of HER-2 spans residues 563-626 that comprises an extensive disulfide-bonding pattern. To delineate the binding region of HER-2, we have designed four synthetic peptides with different levels of conformational flexibility. Chimeric peptides incorporating the measles virus fusion "promiscuous" T cell epitope via a four-residue linker sequence were synthesized, purified, and characterized. All conformational peptides were recognized by trastuzumab and prevented the function of trastuzumab inhibiting tumor cell proliferation, with 563-598 and 597-626 showing greater reactivity. All epitopes were immunogenic in FVB/N mice with Abs against 597-626 and 613-626 recognizing HER-2. The 597-626 epitope was immunogenic in outbred rabbits eliciting Abs which recognized HER-2, competed with trastuzumab for the same epitope, inhibited proliferation of HER-2-expressing breast cancer cells in vitro and caused their Ab-dependent cell-mediated cytotoxicity. Moreover, immunization with the 597-626 epitope significantly reduced tumor burden in transgenic BALB-neuT mice. These results suggest the peptide B cell immunogen is appropriate as a vaccine for HER-2-overexpressing cancers because the resulting Abs show analogous biological properties to trastuzumab.  相似文献   

18.
To study DNA vaccination directed against human HER-2 in the HHD mouse Tg strain, we created a novel HER-2-expressing syngeneic tumor transplantation model. We found that a DNA vaccine encoding the full length HER-2 DNA protected HHD mice from HER-2+ tumor challenge by a CTL independent mechanism. A more efficient approach to induce HLA-A2 restricted CTLs, through immunization with a multi-epitope DNA vaccine expressing the HLA-A2 restricted HER-2 369–377, 435–443 and 689–697 epitopes, resulted in high numbers of peptide specific T cells but failed to induce tumor protection. Subsequently we discovered that HER-2 transfected tumor cells down-regulated MHC class I antigen expression and exhibited a series of defects in the antigen processing pathway which impaired the capacity to produce and display MHC class I peptide-ligands to specific CTLs. Our data demonstrate that HER-2 transfection is associated with defects in the MHC class I presentation pathway, which may be the underlying mechanism behind the inability of CTLs to recognize tumors in this HLA-A2 transgenic model. As defective MHC class I presentation may be a common characteristic of HER-2 expressing tumors, vaccines targeting HER-2 should aim at inducing an integrated immune response where also CD4+ T cells and antibodies are important components. S. Vertuani and C. Triulzi contributed equally to this work.  相似文献   

19.
Schell TD 《Journal of virology》2004,78(4):1751-1762
Mice that express the viral oncoprotein simian virus 40 (SV40) large T antigen (T-Ag) as a transgene provide useful models for the assessment of the state of the host immune response in the face of spontaneous tumor progression. Line SV11 (H2(b)) mice develop rapidly progressing choroid plexus tumors due to expression of full-length T-Ag from the SV40 promoter. In addition, T-Ag expression in the thymus of SV11 mice results in the deletion of CD8(+) T cells specific for the three H2(b)-restricted immunodominant epitopes of T-Ag. Whether CD8(+) T cells specific for the immunorecessive H2-D(b)-restricted epitope V of T-Ag survive negative selection in SV11 mice has not been determined. Immunization of SV11 mice with rVV-ES-V, a recombinant vaccinia virus expressing epitope V as a minigene, resulted in the induction of weak, but reproducible, epitope V-specific cytotoxic T-lymphocyte (CTL) responses. This weak lytic response corresponded with a decreased frequency of epitope V-specific CTL that could be recruited in SV11 mice. In addition, CTL lines derived from rVV-ES-V-immunized SV11 mice had reduced avidities compared to that seen with CTL derived from healthy mice. Despite this initial weak response, significant numbers of epitope V-specific CD8(+) T cells were detected in SV11 mice ex vivo following a priming-boosting approach and these cells demonstrated high avidity for epitope V. The results suggest that low numbers of tumor-reactive CD8(+) T cells with high avidity for epitope V survive negative selection in SV11 mice but can be expanded by specific boosting approaches in the tumor bearing host.  相似文献   

20.
 In order to examine the use of DNA immunization to block tumor growth, we have developed a model system in which a defined 9-amino-acid epitope from the nucleoprotein of influenza virus is used as a surrogate tumor-associated antigen. A mastocytoma cell line of DBA/2 origin (P815) was transfected with a plasmid encoding the minimal H-2Kd-restricted NP(147–155) cytotoxic T lymphocyte (CTL) epitope, pCMV/NPep, to generate the cell line designated P815-NPep. Mice primed and boosted once with a plasmid encoding the full-length NP gene, pCMV/NP, but not with the minigene pCMV/NPep, developed a strong NP(147–155)-specific CTL response within 2 weeks after the boost. When challenged with 104 P815-NPep cells, pCMV/NP-immunized DBA/2 mice were protected from tumor challenge, whereas control mice immunized with the vector backbone rapidly developed lethal tumor. Importantly, the P815-NPep-immune mice were also protected from a subsequent challenge with the untransfected parental tumor P815. By depleting the NP-immune mice of either CD4+ or CD8+ T cells and then challenging with 104 P815-NPep tumor cells, it was determined that the CD8-depleted mice rapidly developed tumors, whereas the CD4-depleted or non-treated mice were protected. These data clearly indicate that intramuscular (i.m.) plasmid DNA immunization can be used to mobilize an effective CD8+ CTL-mediated antitumor response. Received: 8 May 1997 / Accepted: 28 August 1997  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号