Dual Engineering of Human CD8+ T-Cells with WT1-Specific TCR Gene and Chemokine Receptor Gene Transfer Confers Functionally Strengthened Anti-Cancer Reactivity

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4295-4295
Author(s):  
Yukihiro Miyazaki ◽  
Hiroshi Fujiwara ◽  
Toshiki Ochi ◽  
An Jun ◽  
Toshiaki Shirakata ◽  
...  

Abstract Abstract 4295 Background & Purpose: Currently a novel adoptive therapy with engineered T-cells using cancer antigen-specific T-cell receptor (TCR) gene transfer has attracted the attention as a challenging option for cancer treatment. However, reported efficacy from clinical trials using TCR-gene transfer was generally less than expected. In concurrent with the major issue of generation of mispaired TCRs between introduced and endogenous TCR α/β chains, the less accumulation of adoptively infused engineered T-cells at local tumor site in number could also impede the clinical efficacy. Therefore, for the purpose of sufficient accumulation of adoptively transferred tumor-specific T cells into local tumor sites, we set out to examine the feasibility of dual transduction of cancer antigen-specific TCR gene and chemokine receptor gene into human T-cells. Methods: HLA-A*2402-restricted and WT1235-243-specific TCR α/β genes were cloned into a novel GaLV-pseudotyped retroviral vector carrying silencers for endogenous TCR-a/b chains. With retronectin (TakaraBio, Inc.)-coated plate, WT1-specific TCR-α/β genes were introduced into normal CD8+ T-cells with upto more than 50% of WT1-tetramer positivity. On the other hand, using QRT-PCR, we comprehensively screened the chemokine expression profiles of pre-determined HLA-A*2402+ human leukemia cell lines (n=13) and lung cancer cell lines (n=4) which were all WT1-expressing and sensitive to WT1235-243-specific TCR-transduced CD8+ T-cells. Then, the receptor gene for selected chemokine was cloned and inserted into retroviral vector. Expression and function of induced chemokine receptor into Jurkat cells was examined by flowcytometer and transwell experiment. Next, the chemokine receptor gene was introduced into WT1-specific TCR introduced CD8+ T-cells. Migration ability toward target chemokine, and cytotoxicity against the target tumor cells of double-gene transfectans were examined by transwell experiment and 51Cr-releasing assay, respectively. Finally, the combined anti-cancer effect of double-gene transfectants was examined in a novel assay consisted of transwell part and cytotoxicity assay determined by concentration of LDH released from target tumor cells in the bottom well, which were killed by migrated double-gene transfectants from the upper well towards the chemokine produced by target tumor cells. Results: CCL2 chemokine was abundantly produced by some of human lung cancer cell lines and leukemia cell lines. Additionally, receptor for CCL2 (CCR2) was not expressed in activated CD8+ T-cells. Thus, we selected CCL2/CCR2 interaction for this system. CCR2 gene was successfully transduced into Jurkat cells and conferred migration activity towards CCL2 and CCL2 producing lung cnacer cell line LK79 and leukemia cell line KH88. CCR2 gene was also successfully introduced into WT1-specific TCR gene trnasduced CD8+ T-cells. The double-gene transfectants successfully migrated towards CCL2-producing LK79 and KH88 cells. Eventually, in our novel assay system, only double genes-transduced CD8+ T cells in the upper-well, but not single WT1-specific TCR gene-transduced CD8+ T-cells, could migrate towards LK79 cells in the bottom-well and efficiently killed LK79 cells. Background: Although further investigations are warranted, the dual T-cell engineering of human T-cells with chemokine receptor gene and our novel avidity-enhanced tumor-specific TCR gene may be able to more efficiently accumulate engineered T-cells at local tumor sites, which may enhance the immediate anti-tumor effect of adoptively transferred such engineered T-cells. Disclosures: No relevant conflicts of interest to declare.

2003 ◽  
Vol 3 (10) ◽  
pp. 1216-1229 ◽  
Author(s):  
Dongmei Chen ◽  
Yaozhong Ding ◽  
Bernd Schröppel ◽  
Nan Zhang ◽  
Shuang Fu ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4290-4290
Author(s):  
Kozo Nagai ◽  
Hiroshi Fujiwara ◽  
Toshiki Ochi ◽  
Jun An ◽  
Toshiaki Shirakata ◽  
...  

Abstract Abstract 4290 Background & Purpose: Recently we have identified a novel HLA-A*0201-restricted antigenic 9mer epitope (aa207-215: YLILEYAPL) derived from Aurora-A kinase (Aurora-A) which is capable of generating anti-leukemia cytotoxic T-lymphocytes (CTL).(Blood, 2009) To improve the feasibility of Aurora-A targeting cellular immunotherapy against leukemia, we have established a Aurora A207-215-specific CTL clone, and have obtained the full-length T-cell receptor (TCR) α/β genes for TCR gene transfer. In this study, using human leukemia cell lines and patients' leukemia cells, we examined the anti-leukemia reactivity of engineered T-cells with Aurora-A-specific TCR gene transfer. Methods: Full-length of an HLA-A*0201-restricted and Aurora-A207-215-specific TCR α/β genes (Vα3/J20/Cα, Vβ10.3(12)/J1.1/Cβ1, respectively) were cloned into bicistronic GaLV-pseudotyped retroviral vector. Using Retronectin (Takara Bio. Japan)-coated plates, Aurora-A-specific TCR α/β genes were inserted into lymphocytes. Whether this vector was capable of generating a functional Aurora-A207-215-specific TCR heterodimer was examined using Jurkat/MA cells (kindly gifted from Prof. Erik Hooijberg, Netherlands). The epitope-specific and leukemia specific cytotoxicity and IFN-γ production of gene-modified normal CD8+ and CD4+ T-cells were examined by 51Cr-releasing assay and ELISA. HLA-A*0201-restriction of engineered T-cell responses was examined by inhibition assay with antibodies, and HLA-A*0201 transduced human leukemia cell line: MEG01 which abundantly expresses Aurora-A. In vivo anti-leukemia effect of gene-modified CD8+T-cells was examined using NOD/SCID/γcnull (NSG) mice. Eventually, the on-target adverse effect of these Aurora-A-specific TCR-gene transferred CD8+T-cells against autologous hematopoietic progenitor cells was examined using cord blood CD34+ cells. Results: The Aurora-A specific-TCR expressing retroviral vector was capable of generating a functional TCR in Jurkat/MA cells which could produce luciferase in response to Aurora-A peptide on C1R-A2 cells in a dose dependent manner. Aurora-A-specific TCR-transduced CD8+ T-cells produced IFN-γ and exerted cytotoxicity against Aurora-A peptide-loaded C1R-A2 cells in an HLA-A*0201 restricted fashion. These engineered CD8+ T-cells also killed HLA-A*0201+ leukemia cell line and patient leukemia cells, but not HLA-A*0201+ normal PBMC and normal mitotic PHA-stimulated lymphoblasts. The anti-leukemia effect of These engineered CD8+ T cells was significantly abrogated by the anti-HLA-class I monoclonal antibody (MoAb), but not by anti-HLA-DR-MoAb. These engineered CD8+ T-cells killed HLA-A*0201-transduced MEG01 cells which were abundantly expressing Aurora-A, but not parent HLA-A*0201-negative MEG01 cells. Aurora-A-specific TCR gene transduced CD4+ T-cells produced IFN-γ in response to the epitope recognition, which was also in an HLA-A*0201-restricted fashion. Furthermore, Aurora-A-specific TCR-transduced CD8+ T cells did not damage the viability of autologous cord blood CD34+ cells in vitro. Finally, These engineered CD8+ T-cells successfully inhibited the engraftment and growth of inoculated leukemia cell line cells in the NOD/SCIDγ/cnull mice. Background: In this study, Aurora-A kinase-specific TCR gene transferred T-cells successfully recognized the target epitope and exerted the target-specific cytotoxicity. Additionally these engineered CD8+ T-cells exerted anti-leukemia effect both in vitro and in vivo. While those these transfectants did not damage autologous hematopoietic progenitor cells in vitro. Collectively, the novel anti-leukemia adoptive therapy using Aurora-A-specific TCR-gene transferred T-cells appears promising, and further investigations are warranted for the clinical application. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (12) ◽  
pp. 3463-3471 ◽  
Author(s):  
Christoph Hess ◽  
Terry K. Means ◽  
Patrick Autissier ◽  
Tonia Woodberry ◽  
Marcus Altfeld ◽  
...  

CD8 T cells play a key role in host defense against intracellular pathogens. Efficient migration of these cells into sites of infection is therefore intimately linked to their effector function. The molecular mechanisms that control CD8 T-cell trafficking into sites of infection and inflammation are not well understood, but the chemokine/chemokine receptor system is thought to orchestrate this process. Here we systematically examined the chemokine receptor profile expressed on human CD8 T cells. Surprisingly, we found that CXC chemokine receptor 1 (CXCR1), the predominant neutrophil chemokine receptor, defined a novel interleukin-8/CXC ligand 8 (IL-8/CXCL8)–responsive CD8 T-cell subset that was enriched in perforin, granzyme B, and interferon-γ (IFNγ), and had high cytotoxic potential. CXCR1 expression was down-regulated by antigen stimulation both in vitro and in vivo, suggesting antigen-dependent shaping of the migratory characteristics of CD8 T cells. On virus-specific CD8 T cells from persons with a history of Epstein-Barr virus (EBV) and influenza infection, CXCR1 expression was restricted to terminally differentiated effector memory cells. In HIV-1 infection, CXCR1-expressing HIV-1–specific CD8 T cells were present only in persons who were able to control HIV-1 replication during structured treatment interruptions. Thus, CXCR1 identifies a subset of CD8 T cells poised for immediate cytotoxicity and early recruitment into sites of innate immune system activation.


2004 ◽  
Vol 64 (21) ◽  
pp. 7697-7701 ◽  
Author(s):  
Irene M. Mullins ◽  
Craig L. Slingluff ◽  
Jae K. Lee ◽  
Courtney F. Garbee ◽  
Jianfen Shu ◽  
...  

2018 ◽  
Vol 200 (8) ◽  
pp. 2965-2977
Author(s):  
Pedro O. Flores-Villanueva ◽  
Malathesha Ganachari ◽  
Heinner Guio ◽  
Jaime A. Mejia ◽  
Julio Granados

2006 ◽  
Vol 5 (3) ◽  
pp. 98-104
Author(s):  
Yu. Yu. Orlova ◽  
V. M. Alifirova ◽  
N. V. Cherdyntseva ◽  
P. A. Gervas

Multiple sclerosis is chronic inflammatory disease of the central nervous system in the development of which chemokines of the type Tx1 play the leading role. Chemokines and their receptors participate in the development of multiple sclerosis as a result of drawing immune cells into central nervous system. Mutation of CCR5 delta32 decreases functional activity of the appropriate receptor on cellular surface and thus can reduce migration of leucocytes into foci of injury. Aimed at studying the role of mutation in multiple sclerosis, we compared frequency of gene type CCR5 in peripheral mononuclears of 102 multiple sclerosis patients and in 136 healthy subjects. The results obtained allow to conclude that polymorphism of chemokine receptor gene CCR5del32 is not a leading factor in the susceptibility to multiple sclerosis in the studied population.


Blood ◽  
1987 ◽  
Vol 70 (4) ◽  
pp. 1151-1160 ◽  
Author(s):  
E Paietta ◽  
RJ Stockert ◽  
T Calvelli ◽  
P Papenhausen ◽  
SV Seremetis ◽  
...  

A cell line with immature blast cell morphology was isolated from HL-60 promyelocytic leukemia cell cultures and designated HL-T. This new cell type is biphenotypic, expressing terminal transferase (TdT) together with myelomonocytoid immunologic features. TdT enzymatic activity, undetectable in HL-60, was determined to be 140 to 180 units/10(8) HL-T cells by the dGTP-assay, approximately 20% of the activity found in lymphoblastoid cell lines. HL-T predominantly synthesize the known 58- kDa TdT-protein plus a minor 54/56-kDa doublet. The 58-kDa steady state form is nonglycosylated and is phosphorylated. Precursor antigens S3.13 and MY-10, absent on HL-60, are expressed by HL-T; however, the cells are negative for HLA-Dr. Southern blot analysis by hybridization with immunoglobulin heavy chain (JH) and T cell-receptor chain gene (T beta) probes shows JH to be in the germ-line configuration in both cell lines and the T beta gene to be in germ-line in HL-60 but to be rearranged in HL-T. Truncation of the gene encoding the granulocyte-macrophage-colony- stimulating factor (GM-CSF), as found in HL-60, is not observed in HL- T. HL-T are resistant to differentiation-induction by retinoic acid and 1,25-dihydroxyvitamin D3. Cytogenetically HL-T share with HL-60 a deletion of the short arm of chromosome 9 at breakpoint p13, an aberration frequently found in patients with T cell leukemia. In addition, HL-T display t(8;9)(p11;p24) and trisomy 20. Tetraploidy is observed in 80% of HL-T metaphases with aberrations identical to those in the diploid karyotype. Like HL-60, the new line shows some surface- antigenic-T cell characteristics. Despite an antigenic pattern most consistent with that of helper-inducer T cells (T4+, D44+/-, 4B4+, 2H4- , TQ1+/-), HL-T cells and their conditioned culture medium suppress antigen, mitogen, and mixed-leukocyte-culture-mediated lymphocyte proliferation.


2005 ◽  
Vol 5 (3) ◽  
pp. 271-280 ◽  
Author(s):  
Ieda P. Ribeiro ◽  
Carlos G. Schrago ◽  
Esmeralda A. Soares ◽  
Alcides Pissinatti ◽  
Hector N. Seuanez ◽  
...  

2020 ◽  
Vol 41 (7) ◽  
pp. 894-903
Author(s):  
Yunyan Wu ◽  
Meixiang Sang ◽  
Fei Liu ◽  
Jiandong Zhang ◽  
Weijing Li ◽  
...  

Abstract Cancer testis antigens (CTAs) are promising targets for T cell-based immunotherapy and studies have shown that certain CT genes are epigenetically depressed in cancer cells through DNA demethylation. Melanoma-associated antigen A11 (MAGE-A11) is a CTA that is frequently expressed in esophageal cancer and is correlated with a poor esophageal cancer prognosis. Consequently, MAGE-A11 is a potential immunotherapy target. In this study, we evaluated MAGE-A11 expression in esophageal cancer cells and found that it was downregulated in several tumor cell lines, which restricted the effect of immunotherapy. Additionally, the specific recognition and lytic potential of cytotoxic T lymphocytes (CTLs) derived from the MAGE-A11 was determined. Specific CTLs could kill esophageal cancer cells expressing MAGE-A11 but rarely lysed MAGE-A11-negative tumor cells. Therefore, induction of MAGE-A11 expression is critical for CTLs recognition and lysis of esophageal cancer cells. Treatment with the DNA methyltransferase inhibitor 5-aza-2′-deoxycytidine increased MAGE-A11 expression in esophageal cancer cells and subsequently enhanced the cytotoxicity of MAGE-A11-specific CD8+T cells against cancer cell lines. Furthermore, we found that PD-L1 expression in esophageal cancer cells affected the antitumor function of CTLs. programmed death-1 (PD-1)/PD-L1 blockade could increase the specific CTL-induced lysis of HLA-A2+/MAGE-A11+ tumor cell lines treated with 5-aza-2′-deoxycytidine. These findings indicate that the treatment of tumor cells with the DNA methyltransferase inhibitor 5-aza-2′-deoxycytidine augments MAGE-A11 expression in esophageal cancer cells. The combination of epigenetic modulation by 5-aza-2′-deoxycytidine and PD-1/PD-L1 blockade may be useful for T cell-based immunotherapy against esophageal cancer.


Sign in / Sign up

Export Citation Format

Share Document