Network


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

Hotspot


Dive into the research topics where Galit Eisenberg is active.

Publication


Featured researches published by Galit Eisenberg.


Cancer Research | 2008

Capture of Tumor Cell Membranes by Trogocytosis Facilitates Detection and Isolation of Tumor-Specific Functional CTLs

Arthur Machlenkin; Ronny Uzana; Shoshana Frankenburg; Galit Eisenberg; Lea Eisenbach; Jacob Pitcovski; Raphael Gorodetsky; Aviram Nissan; Tamar Peretz; Michal Lotem

The success of adoptive cell transfer in the treatment of metastatic cancer in humans is dependent on the selection of highly active tumor-specific cytotoxic T cells. We report here that CTLs capture membrane fragments from their targets while exerting cytotoxic activity and thus gain a detectable functional signature by which they can be identified. Fluorochrome labeling or biotinylation was used to tag tumor cells. CD8(+) T cells were coincubated with the tagged targets, sorted, and functionally evaluated. Our results show that membrane capture by CD8(+) lymphocytes is T-cell receptor dependent, epitope specific, and preferentially associated with highly cytotoxic clonal subsets. CTLs that captured membranes from unmodified melanoma exhibited enhanced cytotoxic activity against tumor cell lines and autologous melanoma. In a human melanoma in vivo model, adoptive transfer of membrane-capturing, peptide-specific T cells, but not noncapturing or bulk CD8(+) T cells, inhibits tumor progression. Membrane capture is therefore a signature of antigen-specific CTLs endowed with high functional avidity and may have direct relevance in the clinical application of adoptive immunotherapy.


Journal of Immunology | 2013

Imprinting of Lymphocytes with Melanoma Antigens Acquired by Trogocytosis Facilitates Identification of Tumor-Reactive T Cells

Galit Eisenberg; Ronny Uzana; Aviad Pato; Shoshana Frankenburg; Sharon Merims; Eitan Yefenof; Soldano Ferrone; Tamar Peretz; Arthur Machlenkin; Michal Lotem

Trogocytosis is a contact-dependent intercellular transfer of membrane fragments and associated molecules from APCs to effector lymphocytes. We previously demonstrated that trogocytosis also occurs between tumor target and cognate melanoma Ag-specific cytotoxic T cells (CTL). In this study, we show that, following trogocytosis, immune effector cells acquire molecular components of the tumor, including surface Ags, which are detectable by specific mAbs. We demonstrate that CD8+ and CD4+ T cells from melanoma patients’ PBMC and tumor-infiltrating lymphocytes (TIL) capture melanoma Ags, enabling identification of trogocytosing lymphocytes by staining with Ag-specific Abs. This finding circumvents the necessity of tumor prelabeling, which in the past was mandatory to detect membrane-capturing T cells. Through the detection of melanoma Ags on TIL, we sorted trogocytosing T cells and verified their preferential reactivity and cytotoxicity. Furthermore, tumor Ag–imprinted T cells were detected at low frequency in fresh TIL cultures shortly after extraction from the tumor. Thus, T cell imprinting by tumor Ags may allow the enrichment of melanoma Ag-specific T cells for research and potentially even for the adoptive immunotherapy of patients with cancer.


Journal of Immunology | 2012

Trogocytosis Is a Gateway to Characterize Functional Diversity in Melanoma-Specific CD8 + T Cell Clones

Ronny Uzana; Galit Eisenberg; Yael Sagi; Shoshana Frankenburg; Sharon Merims; Ninette Amariglio; Eitan Yefenof; Tamar Peretz; Arthur Machlenkin; Michal Lotem

Trogocytosis, the transfer of membrane patches from target to immune effector cells, is a signature of tumor–T cell interaction. In this study, we used the trogocytosis phenomenon to study functional diversity within tumor-specific T cell clones with identical TCR specificity. MART-126–35–specific CD8 T cell clones, which differed in their trogocytosis capacity (low [2D11], intermediate [2G1], high [2E2]), were generated from melanoma patients. Functional evaluation of the clones showed that the percentage of trogocytosis-capable T cells closely paralleled each clone’s IFN-γ and TNF-α production, lysosome degranulation, and lysis of peptide-pulsed targets and unmodified melanoma. The highly cytotoxic 2E2 clone displayed the highest TCR peptide binding affinity, whereas the low-activity 2D11 clone showed TCR binding to peptide-MHC in a CD8-dependent manner. TCR analysis revealed Vβ16 for clones 2E2 and 2G1 and Vβ14 for 2D11. When peptide-affinity differences were bypassed by nonspecific TCR stimulation, clones 2E2 and 2D11 still manifested distinctive signaling patterns. The high-activity 2E2 clone displayed prolonged phosphorylation of ribosomal protein S6, an integrator of MAPK and AKT activation, whereas the low-activity 2D11 clone generated shorter and weaker phosphorylation. Screening the two clones with identical TCR Vβ by immunoreceptor array showed higher phosphorylation of NK, T, and B cell Ag (NTB-A), a SLAM family homophilic receptor, in clone 2E2 compared with 2G1. Specific blocking of NTB-A on APCs markedly reduced cytokine production by CD8 lymphocytes, pointing to a possible contribution of NTB-A costimulation to T cell functional diversity. This finding identifies NTB-A as a potential target for improving anti-cancer immunotherapy.


Cellular Immunology | 2010

Transcutaneous immunization with hydrophilic recombinant gp100 protein induces antigen-specific cellular immune response.

Galit Eisenberg; Arthur Machlenkin; Shoshana Frankenburg; Adva Mansura; Jacob Pitcovski; Eitan Yefenof; Tamar Peretz; Michal Lotem

The objective of this study was to evaluate the potential of transcutaneous immunization with tumor antigen to induce cell-mediated immunity. For this purpose, hydrophilic recombinant gp100 protein (HR-gp100) was topically applied on human intact skin in vitro, and used as a vaccine in a mouse model. We demonstrate that HR-gp100 permeates into human skin, and is processed and presented by human dendritic cells. In a mouse model, an HR-gp100-based vaccine triggered antigen-specific T cell responses, as shown by proliferation assays, ELISA and intracellular staining for IFN-γ. Transcutaneous antigen delivery may provide a safe, simple and effective method to elicit cell-mediated immunity.


Clinical & Developmental Immunology | 2016

Adjuvant Autologous Melanoma Vaccine for Macroscopic Stage III Disease: Survival, Biomarkers, and Improved Response to CTLA-4 Blockade

Michal Lotem; Sharon Merims; Stephen Frank; Tamar Hamburger; Aviram Nissan; Luna Kadouri; Jonathan Cohen; Ravid Straussman; Galit Eisenberg; Shoshana Frankenburg; Einat Carmon; Bilal Alaiyan; Shlomo Shneibaum; Zeynep Ozge Ayyildiz; Murat Isbilen; Kerem Mert Senses; Ilan G. Ron; Hanna Steinberg; Yoav Smith; Eitan Shiloni; Ali O. Gure; Tamar Peretz

Background. There is not yet an agreed adjuvant treatment for melanoma patients with American Joint Committee on Cancer stages III B and C. We report administration of an autologous melanoma vaccine to prevent disease recurrence. Patients and Methods. 126 patients received eight doses of irradiated autologous melanoma cells conjugated to dinitrophenyl and mixed with BCG. Delayed type hypersensitivity (DTH) response to unmodified melanoma cells was determined on the vaccine days 5 and 8. Gene expression analysis was performed on 35 tumors from patients with good or poor survival. Results. Median overall survival was 88 months with a 5-year survival of 54%. Patients attaining a strong DTH response had a significantly better (p = 0.0001) 5-year overall survival of 75% compared with 44% in patients without a strong response. Gene expression array linked a 50-gene signature to prognosis, including a cluster of four cancer testis antigens: CTAG2 (NY-ESO-2), MAGEA1, SSX1, and SSX4. Thirty-five patients, who received an autologous vaccine, followed by ipilimumab for progressive disease, had a significantly improved 3-year survival of 46% compared with 19% in nonvaccinated patients treated with ipilimumab alone (p = 0.007). Conclusion. Improved survival in patients attaining a strong DTH and increased response rate with subsequent ipilimumab suggests that the autologous vaccine confers protective immunity.


PLOS ONE | 2015

Human T cell crosstalk is induced by tumor membrane transfer.

Ronny Uzana; Galit Eisenberg; Sharon Merims; Shoshana Frankenburg; Aviad Pato; Eitan Yefenof; Roni Engelstein; Tamar Peretz; Arthur Machlenkin; Michal Lotem

Trogocytosis is a contact-dependent unidirectional transfer of membrane fragments between immune effector cells and their targets, initially detected in T cells following interaction with professional antigen presenting cells (APC). Previously, we have demonstrated that trogocytosis also takes place between melanoma-specific cytotoxic T lymphocytes (CTLs) and their cognate tumors. In the present study, we took this finding a step further, focusing on the ability of melanoma membrane-imprinted CD8+ T cells to act as APCs (CD8+T-APCs). We demonstrate that, following trogocytosis, CD8+T-APCs directly present a variety of melanoma derived peptides to fraternal T cells with the same TCR specificity or to T cells with different TCRs. The resulting T cell-T cell immune synapse leads to (1) Activation of effector CTLs, as determined by proliferation, cytokine secretion and degranulation; (2) Fratricide (killing) of CD8+T-APCs by the activated CTLs. Thus, trogocytosis enables cross-reactivity among CD8+ T cells with interchanging roles of effectors and APCs. This dual function of tumor-reactive CTLs may hint at their ability to amplify or restrict reactivity against the tumor and participate in modulation of the anti-cancer immune response.


Journal of Immunotherapy | 2016

Immune Monitoring of Patients Treated With a Whole-Cell Melanoma Vaccine Engineered to Express 4-1BBL.

Roni Engelstein; Sharon Merims; Galit Eisenberg; Jonathan Cohen; Stephen Frank; Tamar Hamburger; Shoshana Frankenburg; Ilan G. Ron; Ruth Isacson; Tal Grenader; Hanna Steinberg; Cyrille J. Cohen; Tamar Peretz; Michal Lotem

CD8 lymphocytes are mandatory mediators of tumor regression. To enhance their specific antitumor activity, we aimed to improve a melanoma cell-based vaccine by transfecting it with 4-1BB ligand, a costimulatory and immune modulatory molecule. Thirty-four American Joint Committee on Cancer (AJCC) stage IIB–IV patients were vaccinated with a melanoma antigen-rich cell line engineered to express HLA-A2 and 4-1BBL (M20/A2/BBL). Twelve serially recruited patients were monitored for interferon &ggr; expression and CD107a mobilization before and after vaccination. Thirty-three patients remained alive, with an estimated mean overall survival of 26.2 months. No grade 3–4 adverse events were encountered. Immune monitoring detected an increase in circulating antimelanoma CD8 T cells in 9 of 12 patients, which were significantly stimulated by the parental melanoma, reflecting a relevant antitumor response. The results from this study show that the costimulatory 4-1BB ligand fortifies an antigen-rich melanoma cell line with enhanced antigen-specific stimulation of CD8 T cells. The use of a costimulatory molecule as part of a vaccine confers a selective increase of T-cell subsets with antimelanoma reactivity, which in some cases were characterized for their epitope specificity.


Clinical and Experimental Immunology | 2015

Messenger RNA encoding constitutively active Toll-like receptor 4 enhances effector functions of human T cells

Aviad Pato; Galit Eisenberg; Arthur Machlenkin; Alon Margalit; G. Cafri; Shoshana Frankenburg; Sharon Merims; Tamar Peretz; Michal Lotem; Gideon Gross

Adoptive T cell therapy of cancer employs a large number of ex‐vivo‐propagated T cells which recognize their targets either by virtue of their endogenous T cell receptor (TCR) or via genetic reprogramming. However, both cell‐extrinsic and intrinsic mechanisms often diminish the in‐vivo potency of these therapeutic T cells, limiting their clinical efficacy and broader use. Direct activation of human T cells by Toll‐like receptor (TLR) ligands induces T cell survival and proliferation, boosts the production of proinflammatory cytokines and augments resistance to regulatory T cell (Treg) suppression. Removal of the TLR ligand‐binding region results in constitutive signalling triggered by the remaining cytosolic Toll/interleukin‐1 receptor (TIR) domain. The use of such TIR domains therefore offers an ideal means for equipping anti‐tumour T cells with the arsenal of functional attributes required for improving current clinical protocols. Here we show that constitutively active (ca)TLR‐4 can be expressed efficiently in human T cells using mRNA electroporation. The mere expression of caTLR‐4 mRNA in polyclonal CD8 and CD4 T cells induced the production of interferon (IFN)‐γ, triggered the surface expression of CD25, CD69 and 4‐1BB and up‐regulated a panel of cytokines and chemokines. In tumour‐infiltrating lymphocytes prepared from melanoma patients, caTLR‐4 induced robust IFN‐γ secretion in all samples tested. Furthermore, caTLR‐4 enhanced the anti‐melanoma cytolytic activity of tumour‐infiltrating lymphocytes and augmented the secretion of IFN‐γ, tumour necrosis factor (TNF)‐α and granulocyte–macrophage colony‐stimulating factor (GM‐CSF) for at least 4 days post‐transfection. Our results demonstrate that caTLR‐4 is capable of exerting multiple T cell‐enhancing effects and can potentially be used as a genetic adjuvant in adoptive cell therapy.


Journal of Immunotherapy | 2016

Spontaneous Activation of Antigen-presenting Cells by Genes Encoding Truncated Homo-Oligomerizing Derivatives of CD40.

Noam Levin; Aviad Pato; Gal Cafri; Galit Eisenberg; Tamar Peretz; Alon Margalit; Michal Lotem; Gideon Gross

The interaction between the CD40 receptor on antigen-presenting cells (APCs) and its trimeric ligand on CD4 T cells is essential for the initiation and progression of the adaptive immune response. Here we undertook to endow CD40 with the capacity to trigger spontaneous APC activation through ligand-independent oligomerization. To this end we exploited the GCN4 yeast transcriptional activator, which contains a leucine zipper DNA-binding motif that induces homophilic interactions. We incorporated GCN4 variants forming homodimers, trimers, or tetramers at the intracellular domain of human and mouse CD40 and replaced the extracellular portion with peptide-&bgr;2m or other peptide tags. In parallel we examined similarly truncated CD40 monomers lacking a GCN4 motif. The oligomeric products appeared to arrange in high–molecular-weight aggregates and were considerably superior to the monomer in their ability to trigger nuclear factor kB signaling, substantiating the anticipated constitutively active (ca) phenotype. Cumulative results in human and mouse APC lines transfected with caCD40 mRNA revealed spontaneous upregulation of CD80, IL-1&bgr;, TNF&agr;, IL-6, and IL-12, which could be further enhanced by caTLR4 mRNA. In mouse bone-marrow–derived dendritic cells caCD40 upregulated CD80, CD86, MHC-II, and IL-12 and in human monocyte–derived dendritic cells it elevated surface CD80, CD83 CD86, CCR7, and HLA-DR. Oligomeric products carrying the peptide-&bgr;2m extracellular portion could support MHC-I presentation of the linked peptide up to 4 days post-mRNA transfection. These findings demonstrate that the expression of a single caCD40 derivative in APCs can exert multiple immunostimulatory effects, offering a new powerful tool in the design of gene-based cancer vaccines.


Journal of Immunology | 2018

Potent Activation of Human T Cells by mRNA Encoding Constitutively Active CD40

Noam Levin; Hadas Weinstein-Marom; Aviad Pato; Orit Itzhaki; Michal J. Besser; Galit Eisenberg; Tamar Peretz; Michal Lotem; Gideon Gross

New strategies for augmenting the actual performance of therapeutic T cells in vivo are needed for improving clinical outcome of adoptive cell therapy. Cumulative findings suggest that CD40 plays an intrinsic role in T cell costimulation. Recently, we demonstrated the ability of truncated, auto-oligomerizing CD40 derivatives to induce strong activation of APCs in a ligand-independent manner. We reasoned that constitutively active CD40 (caCD40) can similarly exert enhancing effects on human antitumor T cells. To test this assumption, we transfected human T cells with in vitro–transcribed caCD40 mRNA. In polyclonal T cells, caCD40 triggered IFN-γ secretion and upregulated CD25 and 4-1BB. In antimelanoma tumor-infiltrating lymphocytes (TILs), caCD40 induced massive production of IFN-γ, exerting a pronounced synergistic effect when coexpressed with constitutively active TLR4 devoid of its extracellular ligand binding. In unselected “young” TILs, caCD40 reproducibly increased surface expression of CD25, OX40, 4-1BB, CD127, and CD28. Three days post-mRNA electroporation of CD8 TILs, caCD40 elevated IFN-γ and TNF-α production and cytolytic activity in the presence of autologous but not HLA-I–mismatched melanoma. Enhanced killing of autologous melanoma by young TILs was observed 4 d posttransfection. These findings suggest that caCD40 can function as a potent T cell adjuvant and provide essential guidelines for similar manipulation of other key members of the TNFR family.

Collaboration


Dive into the Galit Eisenberg's collaboration.

Top Co-Authors

Avatar

Tamar Peretz

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Shoshana Frankenburg

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Michal Lotem

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Arthur Machlenkin

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ronny Uzana

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Aviad Pato

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Eitan Yefenof

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Roni Engelstein

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Michal Lotem

Hebrew University of Jerusalem

View shared research outputs
Researchain Logo
Decentralizing Knowledge