Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
3.
N Engl J Med ; 387(10): 894-904, 2022 09 08.
Article in English | MEDLINE | ID: mdl-36069871

ABSTRACT

BACKGROUND: Antibody-binding of blood dendritic cell antigen 2 (BDCA2), which is expressed exclusively on plasmacytoid dendritic cells, suppresses the production of type I interferon that is involved in the pathogenesis of systemic lupus erythematosus (SLE). The safety and efficacy of subcutaneous litifilimab, a humanized monoclonal antibody that binds to BDCA2, in patients with SLE have not been extensively studied. METHODS: We conducted a phase 2 trial of litifilimab involving participants with SLE. The initial trial design called for randomly assigning participants to receive litifilimab (at a dose of 50, 150, or 450 mg) or placebo administered subcutaneously at weeks 0, 2, 4, 8, 12, 16, and 20, with the primary end point of evaluating cutaneous lupus activity. The trial design was subsequently modified; adults with SLE, arthritis, and active skin disease were randomly assigned to receive either litifilimab at a dose of 450 mg or placebo. The revised primary end point was the change from baseline in the total number of active joints (defined as the sum of the swollen joints and the tender joints) at week 24. Secondary end points were changes in cutaneous and global disease activity. Safety was also assessed. RESULTS: A total of 334 adults were assessed for eligibility, and 132 underwent randomization (64 were assigned to receive 450-mg litifilimab, 6 to receive 150-mg litifilimab, 6 to receive 50-mg litifilimab, and 56 to receive placebo). The primary analysis was conducted in the 102 participants who had received 450-mg litifilimab or placebo and had at least four tender and at least four swollen joints. The mean (±SD) baseline number of active joints was 19.0±8.4 in the litifilimab group and 21.6±8.5 in the placebo group. The least-squares mean (±SE) change from baseline to week 24 in the total number of active joints was -15.0±1.2 with litifilimab and -11.6±1.3 with placebo (mean difference, -3.4; 95% confidence interval, -6.7 to -0.2; P = 0.04). Most of the secondary end points did not support the results of the analysis of the primary end point. Receipt of litifilimab was associated with adverse events, including two cases of herpes zoster and one case of herpes keratitis. CONCLUSIONS: In a phase 2 trial involving participants with SLE, litifilimab was associated with a greater reduction from baseline in the number of swollen and tender joints than placebo over a period of 24 weeks. Longer and larger trials are required to determine the safety and efficacy of litifilimab for the treatment of SLE. (Funded by Biogen; LILAC ClinicalTrials.gov number, NCT02847598.).


Subject(s)
Antibodies, Monoclonal, Humanized , Lectins, C-Type , Lupus Erythematosus, Systemic , Membrane Glycoproteins , Receptors, Immunologic , Adult , Antibodies, Monoclonal, Humanized/therapeutic use , Double-Blind Method , Humans , Lectins, C-Type/immunology , Lupus Erythematosus, Systemic/drug therapy , Membrane Glycoproteins/immunology , Receptors, Immunologic/immunology , Skin Diseases , Treatment Outcome
4.
N Engl J Med ; 387(4): 321-331, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35939578

ABSTRACT

BACKGROUND: Blood dendritic cell antigen 2 (BDCA2) is a receptor that is exclusively expressed on plasmacytoid dendritic cells, which are implicated in the pathogenesis of lupus erythematosus. Whether treatment with litifilimab, a humanized monoclonal antibody against BDCA2, would be efficacious in reducing disease activity in patients with cutaneous lupus erythematosus has not been extensively studied. METHODS: In this phase 2 trial, we randomly assigned adults with histologically confirmed cutaneous lupus erythematosus with or without systemic manifestations in a 1:1:1:1 ratio to receive subcutaneous litifilimab (at a dose of 50, 150, or 450 mg) or placebo at weeks 0, 2, 4, 8, and 12. We used a dose-response model to assess whether there was a response across the four groups on the basis of the primary end point, which was the percent change from baseline to 16 weeks in the Cutaneous Lupus Erythematosus Disease Area and Severity Index-Activity score (CLASI-A; scores range from 0 to 70, with higher scores indicating more widespread or severe skin involvement). Safety was also assessed. RESULTS: A total of 132 participants were enrolled; 26 were assigned to the 50-mg litifilimab group, 25 to the 150-mg litifilimab group, 48 to the 450-mg litifilimab group, and 33 to the placebo group. Mean CLASI-A scores for the groups at baseline were 15.2, 18.4, 16.5, and 16.5, respectively. The difference from placebo in the change from baseline in CLASI-A score at week 16 was -24.3 percentage points (95% confidence interval [CI] -43.7 to -4.9) in the 50-mg litifilimab group, -33.4 percentage points (95% CI, -52.7 to -14.1) in the 150-mg group, and -28.0 percentage points (95% CI, -44.6 to -11.4) in the 450-mg group. The least squares mean changes were used in the primary analysis of a best-fitting dose-response model across the three drug-dose levels and placebo, which showed a significant effect. Most of the secondary end points did not support the results of the primary analysis. Litifilimab was associated with three cases each of hypersensitivity and oral herpes infection and one case of herpes zoster infection. One case of herpes zoster meningitis occurred 4 months after the participant received the last dose of litifilimab. CONCLUSIONS: In a phase 2 trial involving participants with cutaneous lupus erythematosus, treatment with litifilimab was superior to placebo with regard to a measure of skin disease activity over a period of 16 weeks. Larger and longer trials are needed to determine the effect and safety of litifilimab for the treatment of cutaneous lupus erythematosus. (Funded by Biogen; LILAC ClinicalTrials.gov number, NCT02847598.).


Subject(s)
Antibodies, Monoclonal, Humanized , Lectins, C-Type , Lupus Erythematosus, Cutaneous , Membrane Glycoproteins , Receptors, Immunologic , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dose-Response Relationship, Drug , Double-Blind Method , Herpes Zoster/etiology , Humans , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/immunology , Lupus Erythematosus, Cutaneous/drug therapy , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/immunology , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/immunology , Severity of Illness Index , Treatment Outcome
5.
Rheumatology (Oxford) ; 60(11): 5397-5407, 2021 11 03.
Article in English | MEDLINE | ID: mdl-33956056

ABSTRACT

OBJECTIVE: To evaluate the dose-response, efficacy and safety of dapirolizumab pegol (DZP) in patients with SLE. METHODS: Adults with moderately to severely active SLE (SLEDAI-2K score ≥6 and ≥1 BILAG A or ≥2 BILAG B domain scores), receiving stable CS (≤40 mg/day prednisone-equivalent), antimalarial or immunosuppressant drugs were included. Patients with stable LN (proteinuria ≤2 g/day) not receiving high-dose CS or CYC were permitted entry. Randomized patients received placebo or i.v. DZP (6/24/45 mg/kg) and standard-of-care (SOC) treatment every 4 weeks to week 24, after which patients received only SOC to week 48. The primary objective was to establish a dose-response relationship based on week 24 BILAG-Based Composite Lupus Assessment (BICLA) responder rates. RESULTS: All DZP groups exhibited improvements in clinical and immunological outcomes vs placebo at week 24; however, BICLA responder rates did not fit pre-specified dose-response models [best-fitting model (Emax): P = 0.07]. Incidences of serious treatment-emergent adverse events across DZP groups were low and similar to placebo. Following DZP withdrawal, SLEDAI-2K, physician's global assessment (PGA), BILAG, and Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) scores stabilized; BICLA and SLE Responder Index (SRI-4) responder rates declined (likely due to interventions with disallowed escape medications), BILAG flares increased, and immunologic parameters returned towards baseline. CONCLUSIONS: Although the primary objective was not met, DZP appeared to be well tolerated, and patients exhibited improvements across multiple clinical and immunological measures of disease activity after 24 weeks relative to placebo. The potential clinical benefit of DZP warrants further investigation.


Subject(s)
Immunoglobulin Fab Fragments/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Polyethylene Glycols/therapeutic use , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Immunoglobulin Fab Fragments/pharmacology , Lupus Erythematosus, Systemic/immunology , Male , Middle Aged , Polyethylene Glycols/pharmacology , Treatment Outcome
6.
Lupus Sci Med ; 8(1)2021 03.
Article in English | MEDLINE | ID: mdl-33658303

ABSTRACT

OBJECTIVE: Punch biopsy, a standard diagnostic procedure for patients with cutaneous lupus erythematosus (CLE) carries an infection risk, is invasive, uncomfortable and potentially scarring, and impedes patient recruitment in clinical trials. Non-invasive tape sampling is an alternative that could enable serial evaluation of specific lesions. This cross-sectional pilot research study evaluated the use of a non-invasive adhesive tape device to collect messenger RNA (mRNA) from the skin surface of participants with CLE and healthy volunteers (HVs) and investigated its feasibility to detect biologically meaningful differences between samples collected from participants with CLE and samples from HVs. METHODS: Affected and unaffected skin tape samples and simultaneous punch biopsies were collected from 10 participants with CLE. Unaffected skin tape and punch biopsies were collected from 10 HVs. Paired samples were tested using quantitative PCR for a candidate immune gene panel and semi-quantitative immunohistochemistry for hallmark CLE proteins. RESULTS: mRNA collected using the tape device was of sufficient quality for amplification of 94 candidate immune genes. Among these, we found an interferon (IFN)-dominant gene cluster that differentiated CLE-affected from HV (23-fold change; p<0.001) and CLE-unaffected skin (sevenfold change; p=0.002), respectively. We found a CLE-associated gene cluster that differentiated CLE-affected from HV (fourfold change; p=0.005) and CLE-unaffected skin (fourfold change; p=0.012), respectively. Spearman's correlation between per cent area myxovirus 1 protein immunoreactivity and IFN-dominant mRNA gene cluster expression was highly significant (dermis, rho=0.86, p<0.001). In total, skin tape-derived RNA expression comprising both IFN-dominant and CLE-associated gene clusters correlated with per cent area immunoreactivity of some hallmark CLE-associated proteins in punch biopsies from the same lesions. CONCLUSIONS: A non-invasive tape RNA collection technique is a potential tool for repeated skin biomarker measures throughout a clinical trial.


Subject(s)
Lupus Erythematosus, Cutaneous , Cross-Sectional Studies , Female , Healthy Volunteers , Humans , Male , RNA , Skin
7.
J Clin Invest ; 129(3): 1359-1371, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30645203

ABSTRACT

BACKGROUND: Plasmacytoid DCs (pDC) produce large amounts of type I IFN (IFN-I), cytokines convincingly linked to systemic lupus erythematosus (SLE) pathogenesis. BIIB059 is a humanized mAb that binds blood DC antigen 2 (BDCA2), a pDC-specific receptor that inhibits the production of IFN-I and other inflammatory mediators when ligated. A first-in-human study was conducted to assess safety, tolerability, and pharmacokinetic (PK) and pharmacodynamic (PD) effects of single BIIB059 doses in healthy volunteers (HV) and patients with SLE with active cutaneous disease as well as proof of biological activity and preliminary clinical response in the SLE cohort. METHODS: A randomized, double-blind, placebo-controlled clinical trial was conducted in HV (n = 54) and patients with SLE (n = 12). All subjects were monitored for adverse events. Serum BIIB059 concentrations, BDCA2 levels on pDCs, and IFN-responsive biomarkers in whole blood and skin biopsies were measured. Skin disease activity was determined using the Cutaneous Lupus Erythematosus Disease Area and Severity Index Activity (CLASI-A). RESULTS: Single doses of BIIB059 were associated with favorable safety and PK profiles. BIIB059 administration led to BDCA2 internalization on pDCs, which correlated with circulating BIIB059 levels. BIIB059 administration in patients with SLE decreased expression of IFN response genes in blood, normalized MxA expression, reduced immune infiltrates in skin lesions, and decreased CLASI-A score. CONCLUSIONS: Single doses of BIIB059 were associated with favorable safety and PK/PD profiles and robust target engagement and biological activity, supporting further development of BIIB059 in SLE. The data suggest that targeting pDCs may be beneficial for patients with SLE, especially those with cutaneous manifestations. TRIAL REGISTRATION: ClinicalTrials.gov NCT02106897. FUNDING: Biogen Inc.


Subject(s)
Antibodies, Monoclonal , Lectins, C-Type/antagonists & inhibitors , Lupus Erythematosus, Systemic/drug therapy , Membrane Glycoproteins/antagonists & inhibitors , Receptors, Immunologic/antagonists & inhibitors , Skin Diseases/drug therapy , Skin/immunology , Adolescent , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Dendritic Cells/immunology , Dendritic Cells/pathology , Double-Blind Method , Female , Humans , Lectins, C-Type/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Male , Membrane Glycoproteins/immunology , Middle Aged , Plasma Cells/immunology , Plasma Cells/pathology , Receptors, Immunologic/immunology , Skin/pathology , Skin Diseases/immunology , Skin Diseases/pathology
8.
Int Immunol ; 23(1): 17-27, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21123830

ABSTRACT

BACKGROUND: Intranasal administration of high amount of allergen was shown to induce tolerance and to reverse the allergic phenotype. However, mechanisms of tolerance induction via the mucosal route are still unclear. OBJECTIVES: To characterize the therapeutic effects of intranasal application of ovalbumin (OVA) in a mouse model of bronchial inflammation as well as the cellular and molecular mechanisms leading to protection upon re-exposure to allergen. METHODS: After induction of bronchial inflammation, mice were treated intranasally with OVA and re-exposed to OVA aerosols 10 days later. Bronchoalveolar lavage fluid (BALF), T cell proliferation and cytokine secretion were examined. The respective role of CD4(+)CD25(+) and CD4(+)CD25(-) T cells in the induction of tolerance was analysed. RESULTS: Intranasal treatment with OVA drastically reduced inflammatory cell recruitment into BALF and bronchial hyperresponsiveness upon re-exposure to allergen. Both OVA- specific-proliferation of T cells, T(h)1 and T(h)2 cytokine production from lung and bronchial lymph nodes were inhibited. Transfer of CD4(+)CD25(-) T cells, which strongly expressed membrane-bound transforming growth factor ß (mTGFß), from tolerized mice protected asthmatic recipient mice from subsequent aerosol challenges. The presence of CD4(+)CD25(+)(Foxp3(+)) T cells during the process of tolerization was indispensable to CD4(+)CD25(-) T cells to acquire regulatory properties. Whereas the presence of IL-10 appeared dispensable in this model, the suppression of CD4(+)CD25(-)mTGFß(+) T cells in transfer experiments significantly impaired the down-regulation of airways inflammation. CONCLUSION: Nasal application of OVA in established asthma led to the induction of CD4(+)CD25(-)mTGFß(+) T cells with regulatory properties, able to confer protection upon allergen re-exposure.


Subject(s)
Asthma/therapy , CD4-Positive T-Lymphocytes/immunology , Desensitization, Immunologic/methods , Immune Tolerance , Interleukin-2 Receptor alpha Subunit/metabolism , Transforming Growth Factor beta/metabolism , Administration, Intranasal , Allergens/administration & dosage , Allergens/immunology , Animals , Asthma/immunology , Bronchoalveolar Lavage Fluid/immunology , Cell Proliferation , Cytokines/biosynthesis , Drug Evaluation, Preclinical , Lung/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/administration & dosage , Ovalbumin/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology
9.
J Immunol ; 182(8): 5140-51, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19342695

ABSTRACT

Invariant NKT cells (iNKT cells) recognize glycolipid Ags via an invariant TCR alpha-chain and play a central role in various immune responses. Although human CD4(+) and CD4(-) iNKT cell subsets both produce Th1 cytokines, the CD4(+) subset displays an enhanced ability to secrete Th2 cytokines and shows regulatory activity. We performed an ex vivo analysis of blood, liver, and tumor iNKT cells from patients with hepatocellular carcinoma and metastases from uveal melanoma or colon carcinoma. Frequencies of Valpha24/Vbeta11 iNKT cells were increased in tumors, especially in patients with hepatocellular carcinoma. The proportions of CD4(+), double negative, and CD8alpha(+) iNKT cell subsets in the blood of patients were similar to those of healthy donors. However, we consistently found that the proportion of CD4(+) iNKT cells increased gradually from blood to liver to tumor. Furthermore, CD4(+) iNKT cell clones generated from healthy donors were functionally distinct from their CD4(-) counterparts, exhibiting higher Th2 cytokine production and lower cytolytic activity. Thus, in the tumor microenvironment the iNKT cell repertoire is modified by the enrichment of CD4(+) iNKT cells, a subset able to generate Th2 cytokines that can inhibit the expansion of tumor Ag-specific CD8(+) T cells. Because CD4(+) iNKT cells appear inefficient in tumor defense and may even favor tumor growth and recurrence, novel iNKT-targeted therapies should restore CD4(-) iNKT cells at the tumor site and specifically induce Th1 cytokine production from all iNKT cell subsets.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Liver Neoplasms/immunology , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/immunology , Adult , Aged , Aged, 80 and over , Antigens, CD1d/immunology , CD3 Complex/immunology , Cloning, Molecular , Female , HeLa Cells , Health , Humans , Male , Middle Aged
10.
Proc Natl Acad Sci U S A ; 105(10): 3849-54, 2008 Mar 11.
Article in English | MEDLINE | ID: mdl-18319339

ABSTRACT

Human cancer vaccines are often prepared with altered "analog" or "heteroclitic" antigens that have been optimized for HLA class I binding, resulting in enhanced immunogenicity. Here, we take advantage of CpG oligodeoxynucleotides as powerful vaccine adjuvants and demonstrate the induction of high T cell frequencies in melanoma patients, despite the use of natural (unmodified) tumor antigenic peptide. Compared with vaccination with analog peptide, natural peptide induced T cell frequencies that were approximately twofold lower. However, T cells showed superior tumor reactivity because of (i) increased functional avidity for natural antigen and (ii) enhancement of T cell activation and effector function. Thus, novel vaccine formulations comprising potent immune stimulators may allow to circumvent the need for modified antigens and can induce highly functional T cells with precise antigen specificity.


Subject(s)
Antigens, Neoplasm/immunology , Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , Amino Acid Sequence , Antigens, Neoplasm/chemistry , Autoantigens/chemistry , Cancer Vaccines/immunology , Clone Cells , Epitopes/immunology , Granzymes/metabolism , Humans , Interferon-gamma/biosynthesis , Lymphocyte Activation/immunology , MART-1 Antigen , Molecular Sequence Data , Neoplasm Proteins/chemistry , Neoplasm Proteins/immunology , Peptides/chemistry , Peptides/immunology , Perforin/metabolism , Vaccination
11.
Clin Vaccine Immunol ; 14(9): 1108-16, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17634505

ABSTRACT

To monitor antigen-specific CD4+ T cells during a recall immune response to tetanus toxoid (TT), a sequential analysis including ex vivo phenotyping and cytokine flow cytometry, followed by cloning and T-cell-receptor (TCR) spectratyping of cytokine-positive CD4+ T cells, was performed. Grossly, twice as many TT-specific CD4+ T-cell clones, ex vivo derived from the CCR7+/- CD69+ interleukin-2-positive (IL-2+) CD4+ subsets, belonged to the central memory (T(CM); CD62L+ CD27+ CCR7+) compared to the effector memory population (T(EM); CD62L- CD27- CCR7-). After the boost, a predominant expansion of the T(CM) population was observed with more limited variations of the T(EM) population. TCR beta-chain-variable region (BV) spectratyping and sequencing confirmed a large concordance between most frequently expressed BV TCR-CDR3 from ex vivo-sorted CCR7+/- CD69+ IL-2+ CD4+ subsets and BV usage of in vitro-derived TT-specific CD4+ T-cell clones, further demonstrating the highly polyclonal but stable character of the specific recall response to TT. Taken together, ex vivo flow cytometry analysis focused on the CCR7+/- CD69+ IL-2+ CD4+ subsets appears to target the bulk of antigen-specific T cells and to reach an analytical power sufficient to adequately delineate in field trials the profile of the antigen-specific response to vaccine.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Tetanus Toxoid/immunology , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Flow Cytometry/methods , Humans , Immunization, Secondary , Interleukin-2/immunology , Lectins, C-Type , Longitudinal Studies , Lymphocyte Activation , Receptors, Antigen, T-Cell/immunology , Receptors, CCR7/immunology , Tetanus Toxoid/administration & dosage
12.
J Immunol ; 178(6): 3566-74, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17339453

ABSTRACT

Activated CD8 T cells develop cytotoxicity against autologous cells bearing foreign Ags and self/tumor Ags. However, self-specific cytolysis needs to be kept under control to avoid overwhelming immunopathology. After peptide vaccination of melanoma patients, we studied molecular and functional properties of T cell subsets specific for the self/tumor Ag Melan-A/MART-1. Ex vivo analysis revealed three Ag-specific effector memory (EM) populations, as follows: CD28-negative EM (EM28(-)) T cells strongly expressing granzyme/perforin, and two EM28(+) subsets, one with high and the other with low level expression of these cytotoxic proteins. For further functional characterization, we generated 117 stable CD8 T cell clones by ex vivo flow cytometry-based sorting of these subsets. All EM28(-)-derived clones lysed target cells with high efficacy. In contrast, EM28(+)-derived clones were heterogenous, and could be classified in two groups, one with high and the other with low killing capacity, correlating with granzyme/perforin expression. High and low killer phenotypes remained surprisingly stable for several months. However, strongly increased granzyme expression and cytotoxicity were observed after exposure to IL-12. Thus, the data reveal a newly identified subset of CD28(+) conditional killer T cells. Because CD28 can mediate strong costimulatory signals, tight cytotoxicity control, as shown in this study through IL-12, may be particularly important for subsets of T cells expressing CD28.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigens, Neoplasm/immunology , CD28 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-12/pharmacology , Isoantigens/immunology , Melanoma/immunology , Peptide Fragments/immunology , Antigens, Neoplasm/administration & dosage , CD28 Antigens/biosynthesis , CD8-Positive T-Lymphocytes/metabolism , Clone Cells , Female , Granzymes/biosynthesis , Granzymes/immunology , Humans , Immunity, Cellular/drug effects , Isoantigens/administration & dosage , Male , Melanoma/metabolism , Melanoma/therapy , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , Peptide Fragments/administration & dosage , Perforin , Pore Forming Cytotoxic Proteins/biosynthesis , Pore Forming Cytotoxic Proteins/immunology , Time Factors , Vaccination
13.
J Immunol ; 177(2): 1338-48, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16818795

ABSTRACT

Despite major progress in T lymphocyte analysis in melanoma patients, TCR repertoire selection and kinetics in response to tumor Ags remain largely unexplored. In this study, using a novel ex vivo molecular-based approach at the single-cell level, we identified a single, naturally primed T cell clone that dominated the human CD8(+) T cell response to the Melan-A/MART-1 Ag. The dominant clone expressed a high-avidity TCR to cognate tumor Ag, efficiently killed tumor cells, and prevailed in the differentiated effector-memory T lymphocyte compartment. TCR sequencing also revealed that this particular clone arose at least 1 year before vaccination, displayed long-term persistence, and efficient homing to metastases. Remarkably, during concomitant vaccination over 3.5 years, the frequency of the pre-existing clone progressively increased, reaching up to 2.5% of the circulating CD8 pool while its effector functions were enhanced. In parallel, the disease stabilized, but subsequently progressed with loss of Melan-A expression by melanoma cells. Collectively, combined ex vivo analysis of T cell differentiation and clonality revealed for the first time a strong expansion of a tumor Ag-specific human T cell clone, comparable to protective virus-specific T cells. The observed successful boosting by peptide vaccination support further development of immunotherapy by including strategies to overcome immune escape.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cell Differentiation/immunology , Epitopes, T-Lymphocyte/immunology , Immunization, Secondary , Melanoma/immunology , Melanoma/therapy , Neoplasm Proteins/immunology , Antigen Presentation/immunology , Antigens, Neoplasm , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/administration & dosage , Clone Cells , Cytotoxicity Tests, Immunologic , Disease Progression , Epitopes, T-Lymphocyte/blood , Humans , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/immunology , Lymphatic Metastasis/immunology , Lymphatic Metastasis/pathology , Lymphocyte Count , MART-1 Antigen , Melanoma/pathology , Melanoma/secondary , Neoplasm Proteins/blood , Receptors, Antigen, T-Cell/analysis , Receptors, Antigen, T-Cell/blood , Receptors, Antigen, T-Cell/metabolism , Time Factors , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
14.
Eur J Immunol ; 36(7): 1805-14, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16761313

ABSTRACT

The aim of T cell vaccines is the expansion of antigen-specific T cells able to confer immune protection against pathogens or tumors. Although increase in absolute cell numbers, effector functions and TCR repertoire of vaccine-induced T cells are often evaluated, their reactivity for the cognate antigen versus their cross-reactive potential is rarely considered. In fact, little information is available regarding the influence of vaccines on T cell fine specificity of antigen recognition despite the impact that this feature may have in protective immunity. To shed light on the cross-reactive potential of vaccine-induced cells, we analyzed the reactivity of CD8(+) T cells following vaccination of HLA-A2(+) melanoma patients with Melan-A peptide, incomplete Freund's adjuvant and CpG-oligodeoxynucleotide adjuvant, which was shown to induce strong expansion of Melan-A-reactive CD8(+) T cells in vivo. A collection of predicted Melan-A cross-reactive peptides, identified from a combinatorial peptide library, was used to probe functional antigen recognition of PBMC ex vivo and Melan-A-reactive CD8(+) T cell clones. While Melan-A-reactive CD8(+) T cells prior to vaccination are usually constituted of widely cross-reactive naive cells, we show that peptide vaccination resulted in expansion of memory T cells displaying a reactivity predominantly restricted to the antigen of interest. Importantly, these cells are tumor-reactive.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Cross-Priming/immunology , Epitopes, T-Lymphocyte/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Antigens, Neoplasm , Cancer Vaccines/administration & dosage , Cells, Cultured , Humans , MART-1 Antigen , Melanoma/immunology , Melanoma/therapy , Neoplasm Proteins/administration & dosage , Peptide Fragments/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
15.
Immunology ; 113(4): 441-52, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15554922

ABSTRACT

CD45 engagement by monoclonal antibodies on human activated T cells triggers tumour necrosis factor-alpha (TNF-alpha) gene transcription in an epitope-specific manner. To dissect the early signalling events leading to TNF-alpha gene expression, we established that CD45 crosslinking resulted in tyrosine phosphorylation of p56lck, ZAP-70, CD3-zeta, LAT and Vav. This was accompanied by down-regulation of membrane-associated protein tyrosine phosphatase activity in the absence of demonstration of enhanced p56lck, p72syk and ZAP-70 kinase activity, which remained constitutive. These early events eventually triggered an intracellular Ca(2+) rise and phosphoinositide turnover. We conclude that down-regulation of membrane-associated tyrosine phosphatase activity by CD45 extracytoplasmic domain multimerization led, in an epitope-specific fashion, to unopposed tyrosine kinase activity and to the activation of the T-cell receptor/CD3 complex signalling cascade, resulting in TNF-alpha gene expression. This model strongly suggests that CD45 extracytoplasmic tail multimerization may contribute to the modulation T-cell functions.


Subject(s)
Down-Regulation/immunology , Leukocyte Common Antigens/immunology , Protein Tyrosine Phosphatases/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Cells, Cultured , Epitopes, T-Lymphocyte/immunology , Humans , Lymphocyte Activation/immunology , Phosphorylation , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...