Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Leukemia ; 31(10): 2191-2199, 2017 10.
Article in English | MEDLINE | ID: mdl-28202953

ABSTRACT

Impressive results have been achieved by adoptively transferring T-cells expressing CD19-specific CARs with binding domains from murine mAbs to treat B-cell malignancies. T-cell mediated immune responses specific for peptides from the murine scFv antigen-binding domain of the CAR can develop in patients and result in premature elimination of CAR T-cells increasing the risk of tumor relapse. As fully human scFv might reduce immunogenicity, we generated CD19-specific human scFvs with similar binding characteristics as the murine FMC63-derived scFv using human Ab/DNA libraries. CARs were constructed in various formats from several scFvs and used to transduce primary human T-cells. The resulting CD19-CAR T-cells were specifically activated by CD19-positive tumor cell lines and primary chronic lymphocytic leukemia cells, and eliminated human lymphoma xenografts in immunodeficient mice. Certain fully human CAR constructs were superior to the FMC63-CAR, which is widely used in clinical trials. Imaging of cell surface distribution of the human CARs revealed no evidence of clustering without target cell engagement, and tonic signaling was not observed. To further reduce potential immunogenicity of the CARs, we also modified the fusion sites between different CAR components. The described fully human CARs for a validated clinical target may reduce immune rejection compared with murine-based CARs.


Subject(s)
Antigens, CD19/immunology , Burkitt Lymphoma/therapy , Immunotherapy, Adoptive/methods , Recombinant Fusion Proteins/immunology , Single-Chain Antibodies/immunology , T-Lymphocytes/transplantation , Animals , Burkitt Lymphoma/pathology , Cell Line, Tumor , Female , Gene Library , HEK293 Cells , Humans , K562 Cells , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Recombinant Fusion Proteins/genetics , Single-Chain Antibodies/genetics , Species Specificity , Transduction, Genetic , Xenograft Model Antitumor Assays
2.
Clin Pharmacol Ther ; 100(3): 252-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27170467

ABSTRACT

Chimeric antigen receptors (CARs) comprise a tumor-targeting moiety, often in the form of a single chain variable fragment derived from a monoclonal antibody, fused to one or more intracellular T-cell signaling sequences. Lymphodepletion chemotherapy followed by infusion of T cells that are genetically modified to express a CD19-specific CAR is a promising therapy for patients with refractory CD19(+) B-cell malignancies, producing rates of complete remission that are remarkably high in acute lymphoblastic leukemia and encouraging in non-Hodgkin lymphoma and chronic lymphocytic leukemia. Responses are often durable, although additional studies are needed to define the role of CAR-T cell immunotherapy in the context of other treatments. CAR-modified T-cell immunotherapy can be complicated by cytokine release syndrome and neurologic toxicity, which in most cases are manageable and reversible. Here we review recent clinical trial data and discuss issues for the field.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD19/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Antigen, T-Cell/immunology , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , B-Lymphocytes , Clinical Trials as Topic , Humans , Immunotherapy, Adoptive/methods , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , T-Lymphocytes/immunology
3.
Leukemia ; 30(2): 492-500, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26369987

ABSTRACT

Adoptive T-cell therapy with gene-modified T cells expressing a tumor-reactive T-cell receptor or chimeric antigen receptor (CAR) is a rapidly growing field of translational medicine and has shown success in the treatment of B-cell malignancies and solid tumors. In all reported trials, patients have received T-cell products comprising random compositions of CD4(+) and CD8(+) naive and memory T cells, meaning that each patient received a different therapeutic agent. This variation may have influenced the efficacy of T-cell therapy, and complicates comparison of outcomes between different patients and across trials. We analyzed CD19 CAR-expressing effector T cells derived from different subsets (CD4(+)/CD8(+) naive, central memory, effector memory). T cells derived from each of the subsets were efficiently transduced and expanded, but showed clear differences in effector function and proliferation in vitro and in vivo. Combining the most potent CD4(+) and CD8(+) CAR-expressing subsets, resulted in synergistic antitumor effects in vivo. We show that CAR-T-cell products generated from defined T-cell subsets can provide uniform potency compared with products derived from unselected T cells that vary in phenotypic composition. These findings have important implications for the formulation of T-cell products for adoptive therapies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/immunology , Animals , Female , Humans , Immunologic Memory , Mice
4.
Tissue Antigens ; 80(2): 119-25, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22568758

ABSTRACT

Minor histocompatibility (H) antigens are targets of graft-vs-host disease and graft-vs-tumor responses after human leukocyte antigen matched allogeneic hematopoietic stem cell transplantation. Recently, we reported a strategy for genetic mapping of linkage disequilibrium blocks that encoded novel minor H antigens using the large dataset from the International HapMap Project combined with conventional immunologic assays to assess recognition of HapMap B-lymphoid cell line by minor H antigen-specific T cells. In this study, we have constructed and provide an online interactive program and demonstrate its utility for searching for single-nucleotide polymorphisms (SNPs) responsible for minor H antigen generation. The website is available as 'HapMap SNP Scanner', and can incorporate T-cell recognition and other data with genotyping datasets from CEU, JPT, CHB, and YRI to provide a list of candidate SNPs that correlate with observed phenotypes. This method should substantially facilitate discovery of novel SNPs responsible for minor H antigens and be applicable for assaying of other specific cell phenotypes (e.g. drug sensitivity) to identify individuals who may benefit from SNP-based customized therapies.


Subject(s)
Hematopoietic Stem Cell Transplantation , Histocompatibility Testing/methods , Minor Histocompatibility Antigens/immunology , Polymorphism, Single Nucleotide , Software , B-Lymphocytes/immunology , Cell Line , Chromosome Mapping , Data Mining , Genotype , HapMap Project , Humans , Internet , Linkage Disequilibrium , Minor Histocompatibility Antigens/genetics , Phenotype , T-Lymphocytes/immunology , Transplantation, Homologous
5.
J Med Primatol ; 40(2): 88-103, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21044089

ABSTRACT

BACKGROUND: Adoptive immunotherapy with antigen-specific effector T-cell (T(E) ) clones is often limited by poor survival of the transferred cells. We describe here a Macaca nemestrina model for studying transfer of T-cell immunity. METHODS: We derived, expanded, and genetically marked CMV-specific CD8(+) T(E) clones with surface markers expressed on B cells. T(E) cells were adoptively transferred, and toxicity, persistence, retention of introduced cell-surface markers, and phenotype of the persisting T cells were evaluated. RESULTS: CD8(+) T(E) clones were efficiently isolated from distinct memory precursors and gene-marking with CD19 or CD20 permitted in vivo tracking by quantitative PCR. CD19 was a more stable surface marker for tracking cells in vivo and was used to re-isolate cells for functional analysis. Clonally derived CD8(+) T(E) cells differentiated in vivo to phenotypically and functionally heterogeneous memory T-cell subsets. CONCLUSIONS: These studies demonstrate the utility of Macaca nemestrina for establishing principles for T-cell therapeutics applicable to humans.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Immunologic Memory , Immunotherapy, Adoptive , Macaca nemestrina/immunology , Models, Animal , Animals , Antigens, CD19/immunology , Antigens, CD20/immunology , Cell Culture Techniques , Cell Movement , Cell Survival , Cells, Cultured , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/microbiology , Neoplasms/immunology , Neoplasms/therapy , Polymerase Chain Reaction
8.
Proc Natl Acad Sci U S A ; 99(25): 16168-73, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12427970

ABSTRACT

Adoptive T cell therapy, involving the ex vivo selection and expansion of antigen-specific T cell clones, provides a means of augmenting antigen-specific immunity without the in vivo constraints that can accompany vaccine-based strategies. A phase I study was performed to evaluate the safety, in vivo persistence, and efficacy of adoptively transferred CD8+ T cell clones targeting the tumor-associated antigens, MART1MelanA and gp100 for the treatment of patients with metastatic melanoma. Four infusions of autologous T cell clones were administered, the first without IL-2 and subsequent infusions with low-dose IL-2 (at 0.25, 0.50, and 1.0 x 10(6) unitsm(2) twice daily for the second, third, and fourth infusions, respectively). Forty-three infusions of MART1MelanA-specific or gp100-specific CD8+ T cell clones were administered to 10 patients. No serious toxicity was observed. We demonstrate that the adoptively transferred T cell clones persist in vivo in response to low-dose IL-2, preferentially localize to tumor sites and mediate an antigen-specific immune response characterized by the elimination of antigen-positive tumor cells, regression of individual metastases, and minor, mixed or stable responses in 8 of 10 patients with refractory, metastatic disease for up to 21 mo.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/transplantation , Immunotherapy, Adoptive , Melanoma/therapy , Neoplasm Proteins/immunology , T-Lymphocyte Subsets/transplantation , Adult , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Clone Cells/immunology , Clone Cells/transplantation , Female , Graft Survival , Humans , Interleukin-2/therapeutic use , MART-1 Antigen , Male , Melanoma/immunology , Melanoma/secondary , Membrane Glycoproteins/immunology , Middle Aged , Monophenol Monooxygenase/immunology , Recurrence , Safety , T-Lymphocyte Subsets/immunology , Treatment Outcome , gp100 Melanoma Antigen
9.
Tissue Antigens ; 59(4): 293-303, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12135428

ABSTRACT

We have evaluated the utility of genetic linkage analysis to identify genes that encode minor histocompatibility antigens using vaccinia virus vectors as a simple and convenient method for transient expression of class I MHC molecules in lymphoblastoid cell lines. As a test case, we used a CTL clone that recognizes HA-8, a minor histocompatibility antigen encoded by the KIAA0020 gene and presented by HLA-A*0201. EBV-transformed B cell lines from individuals in three large pedigrees from the CEPH reference family collection were infected with a recombinant vaccinia virus vector encoding an HLA-A*0201 transgene, which led to high level expression of the MHC restricting allele HLA-A*0201 on the cell surface. HA-8 expression in the vaccinia-infected target cells was then determined using standard in vitro cytotoxicity assays. Pairwise linkage analysis of the segregation of HA-8 expression in these pedigrees demonstrated that the HA-8 gene was tightly linked with a cluster of marker loci located on the distal portion of chromosome 9p. Analysis of 9p marker haplotypes for individuals in the three families identified several individuals with recombinant haplotypes, and these recombination events were used to refine the precision of the HA-8 gene localization further. The data collectively indicate that the HA-8 gene is localized to a 10.3 cM (corresponding to 3.9 Mb) interval of distal 9p that is thought to encode at least 11 genes, including KIAA0020. These results demonstrate that linkage analysis can be used to map minor histocompatibility genes with high precision and accuracy. Over the next years, refinement and annotation of the human genome sequence will undoubtedly increase the utility of linkage analysis as a tool for identifying minor histocompatibility antigen genes.


Subject(s)
Chromosomes, Human, Pair 9 , HLA-A Antigens/genetics , Lod Score , Minor Histocompatibility Antigens/genetics , Antigens, Surface/genetics , B-Lymphocytes/cytology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Transformed , Chromosome Mapping , Epitopes/genetics , Genetic Markers , HLA-A2 Antigen/genetics , Herpesvirus 4, Human/genetics , Humans , Phenotype , Polymorphism, Genetic
10.
Article in English | MEDLINE | ID: mdl-11722994

ABSTRACT

This article discusses changes in the way hematopoietic stem cell allotransplants may be carried out in the future to treat patients with malignant hematological diseases. Specifically, the focus has shifted away from attempts at eradicating underlying diseases through toxic high-dose chemoradiation therapy towards using the stem cell donor's immune cells for that purpose (allogeneic graft-versus-tumor effect). The non-myeloablative transplant approaches hold promise in reducing the morbidity and mortality associated with conventional high-dose chemoradiation therapy, and they allow allogeneic transplants in elderly or medically infirm patients who are at present not candidates for transplantation. In the future, specific graft-versus-tumor responses may become possible by eliciting donor T cell responses to tumor-associated minor histocompatibility antigens. In Section I, Dr. Rainer Storb describes experimental studies in random-bred dogs that rely on non-cytotoxic immunosuppressive agents to establish stable allografts. Powerful postgrafting immunosuppression, traditionally directed at preventing graft-versus-host disease (GVHD), is also used to overcome host-versus-graft (HVG) reactions, thereby dramatically reducing the need for intensive immunosuppressive conditioning programs. Preclinical canine studies have been translated into the clinical setting for treatment of elderly or medically infirm patients with malignant hematological diseases. The pretransplant conditioning has been reduced to a single dose of 2 Gy total body irradiation (TBI) with or without fludarabine. The lack of toxicity makes it possible for transplants to be conducted in the outpatient setting. Multicenter trials have been initiated, and more than 300 patients have been successfully treated with hematopoietic stem cell grafts both from related and unrelated HLA-matched donors. In Section II, Dr. Richard Champlin describes clinical studies with therapeutic strategies that utilize relatively non-toxic, nonmyeloablative disease-specific preparative regimens incorporating fludarabine, together with other chemotherapeutic agents, to achieve disease suppression and engraftment of allogeneic hematopoietic cells and to allow subsequent infusions of donor lymphocytes. Remissions have been seen in patients with acute myelocytic, chronic myelocytic, chronic lymphocytic, leukemias, lymphomas, and myelomas. In Section III, Dr. Stanley Riddell and colleagues describe studies on isolation of T cells reactive with minor histocompatibility (H) antigens and involved both in GVHD and graft-versus-leukemia (GVL) responses. For example, the gene encoding a novel H-Y antigen in humans has been identified and shown to exhibit restricted tissue expression. Acute myelocytic leukemia stem cells were demonstrated to express the H-Y antigen and additional minor H antigens, and engraftment of such cells in NOD/SCID mice could be selectively prevented by minor antigen-specific T-cell clones. An autosomal encoded human minor H antigen associated with chronic GVHD has been demonstrated. A trial evaluating therapy of relapsed acute myelocytic leukemia or acute lymphoblastic leukemia after allogeneic stem cell transplantation with T-cell clones specific for recipient minor H antigens has been initiated.


Subject(s)
Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/methods , Transplantation Conditioning/methods , Aged , Animals , Antigens, Neoplasm/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/toxicity , Graft vs Host Disease/immunology , Graft vs Tumor Effect/immunology , Humans , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/toxicity , Minor Histocompatibility Antigens/immunology , Transplantation, Homologous/immunology , Transplantation, Homologous/methods
12.
J Immunol ; 166(8): 5250-7, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11290810

ABSTRACT

Due to their potent immunostimulatory capacity, dendritic cells (DC) have become the centerpiece of many vaccine regimens. Immature DC (DCimm) capture, process, and present Ags to CD4(+) lymphocytes, which reciprocally activate DCimm through CD40, and the resulting mature DC (DCmat) loose phagocytic capacity, but acquire the ability to efficiently stimulate CD8(+) lymphocytes. Recombinant vaccinia viruses (rVV) provide a rapid, easy, and efficient method to introduce Ags into DC, but we observed that rVV infection of DCimm results in blockade of DC maturation in response to all activation signals, including CD40L, monocyte-conditioned medium, LPS, TNF-alpha, and poly(I:C), and failure to induce a CD8(+) response. By contrast, DCmat can be infected with rVV and induce a CD8(+) response, but, having lost phagocytic activity, fail to process the Ag via the exogenous class II pathway. To overcome these limitations, we used the CMV protein pp65 as a model Ag and designed a gene containing the lysosomal-associated membrane protein 1 targeting sequence (Sig-pp65-LAMP1) to target pp65 to the class II compartment. DCmat infected with rVV-Sig-pp65-LAMP1 induced proliferation of pp65-specific CD4(+) clones and efficiently induced a pp65-specific CD4(+) response, suggesting that after DC maturation the intracellular processing machinery for class II remains intact for at least 16 h. Moreover, infection of DCmat with rVV-Sig-pp65-LAMP1 resulted in at least equivalent presentation to CD8(+) cells as infection with rVV-pp65. These results demonstrate that despite rVV interference with DCimm maturation, a single targeting vector can deliver Ags to DCmat for the effective simultaneous stimulation of both CD4(+) and CD8(+) cells.


Subject(s)
Antigen Presentation , Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lymphocyte Activation , Membrane Glycoproteins/immunology , Antigen Presentation/genetics , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Viral/genetics , Antigens, Viral/immunology , Antigens, Viral/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Clone Cells , Dendritic Cells/cytology , Dendritic Cells/virology , Down-Regulation/genetics , Down-Regulation/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Immunophenotyping , Lymphocyte Activation/genetics , Lysosomal-Associated Membrane Protein 1 , Lysosomal Membrane Proteins , Lysosomes/genetics , Lysosomes/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism
13.
Curr Opin Immunol ; 13(2): 141-6, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11228405

ABSTRACT

Novel immunologic assays now enable visualization of the antigen-specific response to an extent not previously possible. Assessment of not only the numeric frequency but also the functional properties of individual tumor-specific T cells in the endogenous and manipulated immune response has provided insights that will facilitate the development of immunotherapeutic strategies.


Subject(s)
Antigens, Neoplasm/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Cancer Vaccines/immunology , Chemotaxis, Leukocyte , Cytokines/analysis , Cytokines/immunology , HLA Antigens/immunology , Humans , Immunoglobulins/immunology , Lymphocyte Count , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology
15.
Nat Immunol ; 2(3): 255-60, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11224526

ABSTRACT

NKG2D is an activating receptor that stimulates innate immune responses by natural killer cells upon engagement by MIC ligands, which are induced by cellular stress. Because NKG2D is also present on most CD8alphabeta T cells, it may modulate antigen-specific T cell responses, depending on whether MIC molecules--distant homologs of major histocompatibility complex (MHC) class I with no function in antigen presentation--are induced on the surface of pathogen-infected cells. We found that infection by cytomegalovirus (CMV) resulted in substantial increases in MIC on cultured fibroblast and endothelial cells and was associated with induced MIC expression in interstitial pneumonia. MIC engagement of NKG2D potently augmented T cell antigen receptor (TCR)-dependent cytolytic and cytokine responses by CMV-specific CD28- CD8alphabeta T cells. This function overcame viral interference with MHC class I antigen presentation. Combined triggering of TCR-CD3 complexes and NKG2D induced interleukin 2 production and T cell proliferation. Thus NKG2D functioned as a costimulatory receptor that can substitute for CD28.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus/immunology , Histocompatibility Antigens Class I/immunology , Lymphocyte Activation , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Immunologic/immunology , Cells, Cultured , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/metabolism , Cytotoxicity Tests, Immunologic , Endothelium/metabolism , Endothelium/virology , Fibroblasts/metabolism , Fibroblasts/virology , Histocompatibility Antigens Class I/metabolism , Humans , Interleukin-2/biosynthesis , Lung Diseases, Interstitial/immunology , Lung Diseases, Interstitial/metabolism , NK Cell Lectin-Like Receptor Subfamily K , Receptors, Natural Killer Cell , T-Lymphocytes, Cytotoxic/immunology
16.
J Exp Med ; 193(2): 195-206, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11148223

ABSTRACT

Minor histocompatibility antigens (mHAgs) present a significant impediment to organ and bone marrow transplantation between HLA-identical donor and recipient pairs. Here we report the identification of a new HLA-A*0201-restricted mHAg, HA-8. Designation of this mHAg as HA-8 is based on the nomenclature of Goulmy (Goulmy, E. 1996. Curr. Opin. Immunol. 8:75-81). This peptide, RTLDKVLEV, is derived from KIAA0020, a gene of unknown function located on chromosome 9. Polymorphic alleles of KIAA0020 encode the alternative sequences PTLDKVLEV and PTLDKVLEL. Genotypic analysis demonstrated that the HA-8-specific cytotoxic T lymphocyte (CTL) clone SKH-13 recognized only cells that expressed the allele encoding R at P1. However, when PTLDKVLEV was pulsed onto cells, or when a minigene encoding this sequence was used to artificially translocate this peptide into the endoplasmic reticulum, it was recognized by CTLs nearly as well as RTLDKVLEV. This indicates that the failure of CTLs to recognize cells expressing the PTLDKVLEV-encoding allele of KIAA0020 is due to a failure of this peptide to be appropriately proteolyzed or transported. Consistent with the latter possibility, PTLDKVLEV and its longer precursors were transported poorly compared with RTLDKVLEV by transporter associated with antigen processing (TAP). These studies identify a new human mHAg and provide the first evidence that minor histocompatibility differences can result from the altered processing of potential antigens rather than differences in interaction with the relevant major histocompatibility complex molecule or T cell receptor.


Subject(s)
Antigen Presentation , Minor Histocompatibility Antigens/metabolism , Alleles , Amino Acid Sequence , Base Sequence , Clone Cells , DNA Primers/genetics , Epitopes/chemistry , Epitopes/genetics , Female , Humans , Male , Mass Spectrometry , Minor Histocompatibility Antigens/chemistry , Minor Histocompatibility Antigens/genetics , Molecular Sequence Data , Pedigree , Polymorphism, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
17.
J Virol ; 75(2): 799-808, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134293

ABSTRACT

The in vivo persistence of gene-modified cells can be limited by host immune responses to transgene-encoded proteins. In this study we evaluated in a nonhuman primate model whether the administration of a nonmyeloablative regimen consisting of low-dose total-body irradiation with 200 cGy followed by immunosuppression with mycophenolate mofetil and cyclosporin A for 28 and 35 days, respectively, could be used to facilitate persistence of autologous gene-modified T cells when a transgene-specific immune response had already been established or to induce long-lasting tolerance in unprimed recipients. Two macaques (Macaca nemestrina) received infusions of T cells transduced to express either the enhanced green fluorescent protein and neomycin phosphotransferase genes or the hygromycin phosphotransferase and herpes simplex virus thymidine kinase genes. In the absence of immunosuppression, both macaques developed potent class I major histocompatibility complex-restricted CD8(+) cytotoxic T-lymphocyte (CTL) responses that rapidly eliminated the gene-modified T cells and that persisted long term as memory CTL. Treatment with the nonmyeloablative regimen failed to abrogate preexisting memory CTL responses but interfered with the induction of transgene-specific CTL and facilitated in vivo persistence of gene-modified cells in an unprimed host. However, sustained tolerance to gene-modified T cells was not achieved with this regimen, indicating that further modifications will be required to permit sustained persistence of gene-modified T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunosuppression Therapy , Mycophenolic Acid/analogs & derivatives , T-Lymphocytes , Transfection , Transgenes , Animals , Cyclosporine/pharmacology , Green Fluorescent Proteins , Hematopoietic Stem Cell Transplantation , Immunologic Memory , Immunosuppressive Agents/pharmacology , Kanamycin Kinase/genetics , Kanamycin Kinase/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Macaca nemestrina , Mycophenolic Acid/pharmacology , Retroviridae/genetics , Simplexvirus/enzymology , T-Lymphocytes/immunology , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Transplantation, Autologous , Whole-Body Irradiation
18.
J Exp Med ; 192(11): 1637-44, 2000 Dec 04.
Article in English | MEDLINE | ID: mdl-11104805

ABSTRACT

Current strategies for the immunotherapy of melanoma include augmentation of the immune response to tumor antigens represented by melanosomal proteins such as tyrosinase, gp100, and MART-1. The possibility that intentional targeting of tumor antigens representing normal proteins can result in autoimmune toxicity has been postulated but never demonstrated previously in humans. In this study, we describe a patient with metastatic melanoma who developed inflammatory lesions circumscribing pigmented areas of skin after an infusion of MART-1-specific CD8(+) T cell clones. Analysis of the infiltrating lymphocytes in skin and tumor biopsies using T cell-specific peptide-major histocompatibility complex tetramers demonstrated a localized predominance of MART-1-specific CD8(+) T cells (>28% of all CD8 T cells) that was identical to the infused clones (as confirmed by sequencing of the complementarity-determining region 3). In contrast to skin biopsies obtained from the patient before T cell infusion, postinfusion biopsies demonstrated loss of MART-1 expression, evidence of melanocyte damage, and the complete absence of melanocytes in affected regions of the skin. This study provides, for the first time, direct evidence in humans that antigen-specific immunotherapy can target not only antigen-positive tumor cells in vivo but also normal tissues expressing the shared tumor antigen.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy, Adoptive/adverse effects , Melanocytes/immunology , Melanoma/therapy , Neoplasm Proteins/immunology , Skin Neoplasms/therapy , Vitiligo/immunology , Antigens, Neoplasm/biosynthesis , Female , Humans , Immunotherapy, Adoptive/methods , MART-1 Antigen , Melanocytes/cytology , Melanoma/complications , Melanoma/immunology , Melanoma/pathology , Middle Aged , Neoplasm Proteins/biosynthesis , Skin/cytology , Skin/immunology , Skin/pathology , Skin Neoplasms/complications , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/classification , T-Lymphocytes, Cytotoxic/immunology , Vitiligo/etiology , Vitiligo/pathology
19.
J Virol ; 74(17): 8207-12, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10933734

ABSTRACT

The introduction of genes encoding T-cell receptor (TCR) chains specific for human immunodeficiency virus into T cells of infected patients represents a means to quantitatively and qualitatively improve immunity to the virus. Our results demonstrate that the high level of TCR expression required for physiologic functioning can be reproducibly achieved with retroviral vectors encoding full-length unmodified TCR chains under the control of a strong internal constitutive phosphoglycerate kinase promoter.


Subject(s)
Gene Transfer Techniques , Genes, T-Cell Receptor , HIV-1/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, HIV/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Cloning, Molecular , Epitopes , Genetic Vectors , HIV-1/genetics , Humans , Promoter Regions, Genetic , Receptors, Antigen, T-Cell/genetics , Receptors, HIV/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...