Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lior Carmon is active.

Publication


Featured researches published by Lior Carmon.


European Journal of Immunology | 1999

MHC class I-restricted epitope spreading in the context of tumor rejection following vaccination with a single immunodominant CTL epitope

Khaled M. El-Shami; Boaz Tirosh; Erez Bar-Haim; Lior Carmon; Ezra Vadai; Mati Fridkin; Michael Feldman; Lea Eisenbach

Epitope spreading is a process whereby epitopes distinct from and non‐cross‐reactive with an inducing epitope become targets of an evolving immune response. This phenomenon has been associated most notably with the progression of naturally occurring or experimentally induced chronic autoimmune diseases. We have investigated the potential occurrence of epitope spreading in the context of antitumor cytotoxic T cell (CTL) responses using chicken ovalbumin (OVA) as a model antigen. Our results indicate that following rejection of OVA‐expressing EG.7 tumor cells effectuated by a CTL response which is induced against the MHC class I‐restricted immunodominant epitope OVA257 – 264, there occurs intramolecular diversification of the CTL response to two additional OVA‐derived epitopes, OVA176 – 183 and OVA55 – 62, as well as intermolecular spreading to other endogenous tumor‐derived determinants. It seems that CTL‐mediated tumor cell destruction in vivo favors cross‐presentation of additional epitopes with the consequent activation of additional tumor‐reactive lymphocytes. The process of epitope spreading in that context has obvious important implications for the design of antigen‐specific antitumor immunotherapies.


International Journal of Cancer | 2000

Novel breast‐tumor‐associated MUC1‐derived peptides: Characterization in Db−/− × β2 microglobulin (β2m) null mice transgenic for a chimeric HLA‐A2.1/Db‐β2 microglobulin single chain

Lior Carmon; Khaled M. El-Shami; Adrian Paz; Steve Pascolo; Esther Tzehoval; Boaz Tirosh; Romelin Koren; Michael Feldman; Mati Fridkin; François A. Lemonnier; Lea Eisenbach

The MUC1 protein was found to be up‐regulated in a spectrum of malignant tumors. T‐cell responses to the MUC1 extracellular tandem repeat array (TRA) were observed in murine models as well as in breast‐carcinoma patients. In the present study, we evaluated the anti‐tumor potential of HLA‐A2.1‐motif‐selected peptides from non‐TRA domains of the molecule. Peptide immunogenicity was examined in the Db−/− × β2 microglobulin (β2m) null mice transgenic for a modified HLA‐A2.1/Db‐β2 microglobulin single chain (HHD mice). Our results show the existence of 3 novel HLA‐A2.1‐restricted MUC1‐derived cytotoxic T‐lymphocyte (CTL) epitopes. These peptides are processed and presented by the HHD‐transfected breast‐tumor cell line MDA‐MB‐157. Moreover, CTL induced by these 3 peptides show higher lysis of target cells pulsed with breast‐carcinoma‐derived peptides than of targets pulsed with normal breast‐tissue‐derived peptides. These data suggest an important role for non‐TRA MUC1‐derived peptides as inducers of a MHC‐restricted CTL reaction to a breast‐carcinoma cell line and patient‐derived tumor extracts. Int. J. Cancer 85:391–397, 2000. ©2000 Wiley‐Liss, Inc.


Journal of Clinical Investigation | 2002

Characterization of novel breast carcinoma–associated BA46-derived peptides in HLA-A2.1/Db-β2mtransgenic mice

Lior Carmon; Irene Bobilev-Priel; Baruch Brenner; Dimitry Bobilev; Adrian Paz; Erez Bar-Haim; Boaz Tirosh; Tirza Klein; Mati Fridkin; François A. Lemonnier; Esther Tzehoval; Lea Eisenbach

The human milk fat globule membrane protein BA46 (lactadherin) is highly overexpressed in human breast tumors, making it a potential target for tumor immunotherapy. We have identified BA46-derived peptides that contain the motif recognized by the MHC class I molecule HLA-A2.1 and that are processed and presented by human breast carcinoma cells. In mice lacking normal class I molecules but expressing an HLA-A2.1/D(b)-beta2 microglobulin single chain (HHD mice), three peptides elicited specific CTL activity. Two of these peptides also stimulated cytotoxic activity in peripheral blood lymphocytes from HLA-A2.1-positive breast carcinoma patients. Adoptive transfer of HHD-derived bulk CTLs to nude mice bearing human breast carcinoma transplants reduced tumor growth. These peptides therefore represent naturally processed BA46-derived CTL epitopes that can be used in peptide-based antitumor vaccines.


Vaccine | 2011

ImMucin: A novel therapeutic vaccine with promiscuous MHC binding for the treatment of MUC1­expressing tumors

Riva Kovjazin; Ilan Volovitz; Yulia Kundel; Eli Rosenbaum; Gal Medalia; Galit Horn; Nechama I. Smorodinsky; Baruch Brenner; Lior Carmon

An optimal cancer vaccine should be able to induce highly potent, long-lasting, tumor-specific responses in the majority of the cancer patient population. One approach for achieving this is to use synthetic peptide vaccines derived from widely expressed tumor-associated antigens, that promiscuously bind multiple MHC class I and class II alleles. MUC1-SP-L (ImMucin, VXL100) is a 21mer peptide encoding the complete signal peptide domain of MUC1, a tumor-associated antigen expressed by over 90% of solid and non-solid tumors. MUC1-SP-L was predicted in silico to bind various MHC class I and MHC class II alleles, covering the majority of the Caucasian population. PBLs obtained from 13 naïve donors all proliferated, with a Stimulation Index (SI≥2), to the MUC1-SP-L peptide, producing mixed CD4+ and CD8+ responses. Similar results were manifested by MUC1-SP-L in PBLs derived from 9 of 10 cancer patients with MUC1 positive tumors. CD4+ and CD8+ T cell populations exhibited CD45RO memory markers and secreted IFN-gamma and IL-2 following stimulation with MUC1-SP-L. These T cells also exhibited proliferation to the MUC1-SP-L inner 9mer epitopes and cytotoxicity against tumor cell lines expressing MUC1 and a concordant MHC class I allele. Cytotoxicity to MUC1-expressing human and murine tumors was shown also in T cells obtained from HLA-A2 transgenic mice and BALB/c syngeneic mice immunized with the MUC1-SP-L and GM-CSF. In an immunotherapy model, BALB/c mice inoculated with metastatic MUC1 transfected murine DA3 mammary tumor cells, exhibited significantly prolonged survival following vaccination with MUC1-SP-L. Our results indicate superior immunological and anti-tumor properties of MUC1-SP-L compared to previously published MUC1-derived epitopes.


Molecular Immunology | 2011

Signal peptides and trans-membrane regions are broadly immunogenic and have high CD8+ T cell epitope densities: Implications for vaccine development

Riva Kovjazin; Ilan Volovitz; Yair Daon; Tal Vider-Shalit; Roy Azran; Lea Tsaban; Lior Carmon; Yoram Louzoun

Cell mediated immune response has a major role in controlling the elimination of infectious agents. The rational design of sub-unit peptide vaccines against intracellular pathogens or cancer requires the use of antigenic sequence/s that can induce highly potent, long lasting and antigen-specific responses in the majority of the population. A promising peptide selection strategy is the detection of multi-epitope peptide sequences with an ability to bind multiple MHC alleles. While past research sought the best epitopes based on their specific antigenicity, we ask whether specific defined domains have high epitope densities. Signal peptides and trans-membrane domains were found to have exceptionally high epitope densities. The improved MHC binding of these domains relies on their hydrophobic nature and, in signal peptides, also on their specific sequence. The high epitope density of SP was computed using in-silico methods and corroborated by the high percentage of identified SP epitope in the IEDB (immune epitope database). The enhanced immunogenicity of SP was then experimentally confirmed using a panel of nine peptides derived from Mycobacterium tuberculosis (MTb) proteins used in human PBMC proliferation assays and T cell lines functional assays. Our results show the exceptionally high antigen specific response rates and population coverage to SP sequences compared with non-SP peptide antigens derived from the same proteins. The results suggest a novel scheme for the rational design of T cell vaccines using a domain based rather than an epitope based approach.


International Journal of Cancer | 2000

Induction of antitumor immunity by proteasome-inhibited syngeneic fibroblasts pulsed with a modified TAA peptide.

Khaled M. El-Shami; Boaz Tirosh; Dan Popovic; Lior Carmon; Esther Tzehoval; Ezra Vadai; Michael Feldman; Lea Eisenbach

CTLs specific for tumor antigens play a major role in immunity against cancer. Improved binding affinity of putative TAA peptides could enhance the in vivo immunogenicity of these self‐altered self‐ tumor antigens. We examined here the efficacy of tumor vaccines composed of an altered peptide ligand of MUT‐1, designated MUT‐D, which exhibited significantly higher class‐I allele Kb binding affinity than its native counterpart MUT‐1. The peptide was loaded on antigen presenting cells composed of the C57BL/6‐syngeneic fibroblast cell line BLK.CL4. These cells were treated with proteasome inhibitor in order to shut off the degradation of proteins and the subsequent loading of endogenous peptides onto MHC class‐I molecules, thus allowing for the pulsing of these cells with the modified peptide MUT‐D. Proteasome‐inhibited and modified peptide‐loaded fibroblasts induced a peptide‐specific CTL that significantly delayed primary tumor progression and protected the pre‐immunized mice against the development of lung metastasis following the surgical removal of the primary tumor. Genetic modification of the fibroblasts to express the immunostimulatory cytokine IL‐2 did not improve the APC function of the modified cells, nor did it result in augmentation of the potency of the vaccine. Our results suggest that the proteasome‐inhibited fibroblasts pulsed with modified, high binder tumor‐associated antigen peptide are good antigen‐presenting cells and represent an effective form of tumor vaccine. Int. J. Cancer 85:236–242, 2000. ©2000 Wiley‐Liss, Inc.


PLOS ONE | 2014

Cell Surface-Associated Anti-MUC1-Derived Signal Peptide Antibodies: Implications for Cancer Diagnostics and Therapy

Riva Kovjazin; Galit Horn; Nechama I. Smorodinsky; Michael Y. Shapira; Lior Carmon

The MUC1 tumor associated antigen is highly expressed on a range of tumors. Its broad distribution on primary tumors and metastases renders it an attractive target for immunotherapy. After synthesis MUC1 is cleaved, yielding a large soluble extracellular alpha subunit containing the tandem repeats array (TRA) domain specifically bound, via non-covalent interaction, to a smaller beta subunit containing the transmembrane and cytoplasmic domains. Thus far, inconclusive efficacy has been reported for anti-MUC1 antibodies directed against the soluble alpha subunit. Targeting the cell bound beta subunit, may bypass limitations posed by circulating TRA domains. MUC1’s signal peptide (SP) domain promiscuously binds multiple MHC class II and Class I alleles, which upon vaccination, generated robust T-cell immunity against MUC1-positive tumors. This is a first demonstration of non-MHC associated, MUC1 specific, cell surfaces presence for MUC1 SP domain. Polyclonal and monoclonal antibodies generated against MUC1 SP domain specifically bind a large variety of MUC1-positive human solid and haematological tumor cell lines; MUC1-positive bone marrow derived plasma cells obtained from multiple myeloma (MM)-patients, but not MUC1 negative tumors cells, and normal naive primary blood and epithelial cells. Membranal MUC1 SP appears mainly as an independent entity but also co-localized with the full MUC1 molecule. MUC1-SP specific binding in BM-derived plasma cells can assist in selecting patients to be treated with anti-MUC1 SP therapeutic vaccine, ImMucin. A therapeutic potential of the anti-MUC1 SP antibodies was suggested by their ability to support of complement-mediated lysis of MUC1-positive tumor cells but not MUC1 negative tumor cells and normal naive primary epithelial cells. These findings suggest a novel cell surface presence of MUC1 SP domain, a potential therapeutic benefit for anti-MUC1 SP antibodies in MUC1-positive tumors and a selection tool for MM patients to be treated with the anti-MUC1 SP vaccine, ImMucin.


Immunology Letters | 1999

IMMUNOGENICITY OF H-2KB-LOW AFFINITY, HIGH AFFINITY, AND COVALENTLY-BOUND PEPTIDES IN ANTI-TUMOR VACCINATION

Boaz Tirosh; Khaled M. El-Shami; Nora Vaisman; Lior Carmon; Erez Bar-Haim; Ezra Vadai; Michael Feldman; Mati Fridkin; Lea Eisenbach

CTL induction by immunization with synthetic peptide epitopes has been shown to inhibit tumor growth and its metastatic spread. Ex vivo pulsing of peptides on MHC class I-bearing cells such as RMA-S cells or professional APCs elicits an effective CTL response. Since the stability of the MHC-peptide complex is strongly correlated with the overall immunogenecity, we compared the effect of immunization with low affinity, high affinity, and irreversibly bound MHC peptides in the context of immunotherapy of metastasis. MUT1, a tumor-associated antigen peptide that was isolated from 3LL Lewis lung carcinoma, is a low H-2Kb binder. MUT1 was modified into a high binder by changing positions 3, 5, and 8 to the favorable anchor residues. In addition, we introduced a photo-active chemical moiety, which can bind irreversibly to MHC upon illumination. These peptides, loaded onto RMA-S, were used to immunize mice against the 3LL tumor. Vaccination via the covalent conjugation of the low binder peptide was found to increase the CTL response measured against MUT1 loaded cells and against H-2Kb transfected D122 cells relative to the native MUT1 peptide. However, the photo cross-linking of the high affinity peptide to the MHC did not significantly improve the induction of specific CTL. The level of CTL activity was elevated to the same extent by either cross-linking the peptide to the MHC or by modifying it into a high-binder peptide. The protective capacity of all the peptide-based vaccines against D122 metastatic spread to the lungs was found to be comparable. These results indicate that augmentation of the affinity of a TAA peptide to the RMA-S surface MHC molecules, by conversion to a high-affinity mimotope or by photo-conjugation, can significantly enhance the immune response. There seems to be, however, a ceiling beyond which increase in the peptide-binding affinity does not lead to a corresponding enhancement of the overall immunogenicity of the peptide.


Clinical and Vaccine Immunology | 2013

Characterization of Novel Multiantigenic Vaccine Candidates with Pan-HLA Coverage against Mycobacterium tuberculosis

Riva Kovjazin; David Shitrit; Rachel Preiss; Ilanit Haim; Lev Triezer; Leonardo Fuks; Abdel Rahman Nader; Meir Raz; Ritta Bardenstein; Galit Horn; Nechama I. Smorodinsky; Lior Carmon

ABSTRACT The low protection by the bacillus Calmette-Guérin (BCG) vaccine and existence of drug-resistant strains require better anti-Mycobacterium tuberculosis vaccines with a broad, long-lasting, antigen-specific response. Using bioinformatics tools, we identified five 19- to 40-mer signal peptide (SP) domain vaccine candidates (VCs) derived from M. tuberculosis antigens. All VCs were predicted to have promiscuous binding to major histocompatibility complex (MHC) class I and II alleles in large geographic territories worldwide. Peripheral mononuclear cells (PBMC) from healthy naïve donors and tuberculosis patients exhibited strong proliferation that correlated positively with Th1 cytokine secretion only in healthy naïve donors. Proliferation to SP VCs was superior to that to antigen-matched control peptides with similar length and various MHC class I and II binding properties. T-cell lines induced to SP VCs from healthy naïve donors had increased CD44high/CD62L+ activation/effector memory markers and gamma interferon (IFN-γ), but not interleukin-4 (IL-4), production in both CD4+ and CD8+ T-cell subpopulations. T-cell lines from healthy naïve donors and tuberculosis patients also manifested strong, dose-dependent, antigen-specific cytotoxicity against autologous VC-loaded or M. tuberculosis-infected macrophages. Lysis of M. tuberculosis-infected targets was accompanied by high IFN-γ secretion. Various combinations of these five VCs manifested synergic proliferation of PBMC from selected healthy naïve donors. Immunogenicity of the best three combinations, termed Mix1, Mix2, and Mix3 and consisting of 2 to 5 of the VCs, was then evaluated in mice. Each mixture manifested strong cytotoxicity against M. tuberculosis-infected macrophages, while Mix3 also manifested a VC-specific humoral immune response. Based on these results, we plan to evaluate the protection properties of these combinations as an improved tuberculosis subunit vaccine.


Human Vaccines & Immunotherapeutics | 2014

The use of signal peptide domains as vaccine candidates

Riva Kovjazin; Lior Carmon

Signal peptide (SP) domains have a common motif but also sequence specific features. This knowledge was mainly ignored by immunologists who considered SP as generic, short-lived, targeting sequences. Consequently, while SP-derived MHC class I, class II and HLA-E epitopes have been isolated, their use as antigen-specific vaccine candidates (VCs) was mostly neglected. Recently we demonstrated the rational of selecting entire SP domains as multi-epitope long peptide VCs based on their high T and B-cell epitope densities. This review summarizes preclinical and clinical results demonstrating the various advantages of human SP domain VCs derived from both bacterial and tumor antigens. Such vaccine design provides for a straightforward, yet unique immunotherapeutic means of generating robust, non-toxic, diversified, combined antigen-specific CD4+/CD8+ T/B-cell immunity, irrespective of patient HLA repertoire also in disease associated transporter-associated with antigen processing (TAP) deficiencies. Subsequent clinical trials will further assess the full potential of this approach.

Collaboration


Dive into the Lior Carmon's collaboration.

Top Co-Authors

Avatar

Lea Eisenbach

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Boaz Tirosh

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Esther Tzehoval

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Riva Kovjazin

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Adrian Paz

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Khaled M. El-Shami

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Erez Bar-Haim

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Ezra Vadai

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Mati Fridkin

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Michael Feldman

Weizmann Institute of Science

View shared research outputs
Researchain Logo
Decentralizing Knowledge