Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Gut ; 61(8): 1163-71, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22207629

ABSTRACT

BACKGROUND: There is indirect evidence that T cell responses can control the metastatic spread of colorectal cancer (CRC). However, an enrichment of CD4(+)Foxp3(+) regulatory T cells (Tregs) has also been documented. OBJECTIVE: To evaluate whether CRC promotes Treg activity and how this influences anti-tumour immune responses and disease progression. METHODS: A longitudinal study of Treg activity on a cohort of patients was performed before and after tumour resection. Specific CD4(+) T cell responses were also measured to the tumour associated antigens carcinoembryonic antigen (CEA) and 5T4. RESULTS: Tregs from 62 preoperative CRC patients expressed a highly significant increase in levels of Foxp3 compared to healthy age-matched controls (p=0.007), which returned to normal after surgery (p=0.0075). CD4(+) T cell responses to one or both of the tumour associated antigens, CEA and 5T4, were observed in approximately two-thirds of patients and one third of these responses were suppressed by Tregs. Strikingly, in all patients with tumour recurrence at 12 months, significant preoperative suppression was observed of tumour-specific (p=0.003) but not control CD4(+) T cell responses. CONCLUSION: These findings demonstrate that the presence of CRC drives the activity of Tregs and accompanying suppression of CD4(+) T cell responses to tumour-associated antigens. Suppression is associated with recurrence of tumour at 12 months, implying that Tregs contribute to disease progression. These findings offer a rationale for the manipulation of Tregs for therapeutic intervention.


Subject(s)
Adenocarcinoma/immunology , CD4-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Immunity, Cellular , T-Lymphocytes, Regulatory/immunology , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Colectomy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Disease Progression , Female , Flow Cytometry , Follow-Up Studies , Forkhead Transcription Factors/biosynthesis , Humans , Immunohistochemistry , Lymphocyte Activation/immunology , Male , Middle Aged , Neoplasm Recurrence, Local , Neoplasm Staging , Prognosis , Time Factors
2.
Nat Med ; 29(1): 104-114, 2023 01.
Article in English | MEDLINE | ID: mdl-36624315

ABSTRACT

Affinity-optimized T cell receptors can enhance the potency of adoptive T cell therapy. Afamitresgene autoleucel (afami-cel) is a human leukocyte antigen-restricted autologous T cell therapy targeting melanoma-associated antigen A4 (MAGE-A4), a cancer/testis antigen expressed at varying levels in multiple solid tumors. We conducted a multicenter, dose-escalation, phase 1 trial in patients with relapsed/refractory metastatic solid tumors expressing MAGE-A4, including synovial sarcoma (SS), ovarian cancer and head and neck cancer ( NCT03132922 ). The primary endpoint was safety, and the secondary efficacy endpoints included overall response rate (ORR) and duration of response. All patients (N = 38, nine tumor types) experienced Grade ≥3 hematologic toxicities; 55% of patients (90% Grade ≤2) experienced cytokine release syndrome. ORR (all partial response) was 24% (9/38), 7/16 (44%) for SS and 2/22 (9%) for all other cancers. Median duration of response was 25.6 weeks (95% confidence interval (CI): 12.286, not reached) and 28.1 weeks (95% CI: 12.286, not reached) overall and for SS, respectively. Exploratory analyses showed that afami-cel infiltrates tumors, has an interferon-γ-driven mechanism of action and triggers adaptive immune responses. In addition, afami-cel has an acceptable benefit-risk profile, with early and durable responses, especially in patients with metastatic SS. Although the small trial size limits conclusions that can be drawn, the results warrant further testing in larger studies.


Subject(s)
Antigens, Neoplasm , Head and Neck Neoplasms , Male , Humans , Neoplasm Proteins , HLA-A Antigens , Cell- and Tissue-Based Therapy , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods
3.
J Immunol ; 182(9): 5203-7, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380765

ABSTRACT

CD59, a broadly expressed GPI-anchored molecule, regulates formation of the membrane attack complex of the complement cascade. We previously demonstrated that mouse CD59 also down-modulates CD4(+) T cell activity in vivo. In this study, we explored the role of CD59 on human CD4(+) T cells. Our data demonstrate that CD59 is up-regulated on activated CD4(+) T cells and serves to down-modulate their activity in response to polyclonal and Ag-specific stimulation. The therapeutic potential of this finding was explored using T cells isolated from colorectal cancer patients. The findings were striking and indicated that blockade of CD59 significantly enhanced the CD4(+) T cell response to two different tumor Ags. These data highlight the potential for manipulating CD59 expression on T cells for boosting weak immune responses, such as those found in individuals with cancer.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD59 Antigens/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Epitopes, T-Lymphocyte/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/pathology , CD59 Antigens/genetics , CD59 Antigens/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunotherapy, Adoptive , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , U937 Cells , Up-Regulation/genetics , Up-Regulation/immunology
4.
J Allergy Clin Immunol ; 121(5): 1277-1284.e3, 2008 May.
Article in English | MEDLINE | ID: mdl-18355912

ABSTRACT

BACKGROUND: Data from rodent models suggest that a subpopulation of CD4+ T cells, characterized by the constitutive expression of CD25, play a key role in regulating many immune responses. Human CD4+CD25+ T cells also appear to possess a regulatory function, but their role in infections is not fully defined. OBJECTIVES: We sought to explore the possibility of a role for CD4+CD25+ T cells in controlling immunity to hepatitis C virus (HCV). We hypothesized that CD4+CD25+ T cells might account for the paucity of immune responses measurable in chronically viremic patients by suppressing the immune responses to HCV antigens. METHODS: We compared the responses of PBMCs to 3 different recombinant HCV antigens before and after depletion of CD25+ cells in 15 chronically viremic patients, 14 nonviremic HCV antibody-positive subjects, and 14 healthy control subjects. We also tested the ability of CD4+CD25+ T cells purified from HLA-matched viremic or nonviremic blood to suppress the responses of HCV epitope-specific T-cell clones. RESULTS: To our surprise, depletion of peripheral blood CD25+ cells led to a pronounced increase in proliferation of and IFN-gamma production by PBMCs only in nonviremic patients. Furthermore, the CD4+CD25+ T cells purified from HLA-matched nonviremic blood (in contrast to CD4+CD25+ T cells isolated from chronically viremic blood) inhibited the responses of HCV epitope-specific T-cell clones. CONCLUSION: HCV-specific CD4+CD25+ regulatory T cells appear to accompany successful viral clearance.


Subject(s)
Hepatitis C, Chronic/immunology , Hepatitis C/immunology , Hepatitis C/virology , T-Lymphocytes, Regulatory/immunology , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Hepatitis C Antibodies/blood , Hepatitis C Antibodies/immunology , Hepatitis C Antigens/immunology , Humans , Viral Load , Viremia/immunology
5.
Front Immunol ; 10: 889, 2019.
Article in English | MEDLINE | ID: mdl-31068951

ABSTRACT

Regulatory T cells (Tregs) can control excessive or undesirable immune responses toward autoantigens, alloantigens, and pathogens. In transplantation, host immune responses against the allograft are suppressed through the use of immunosuppressive drugs, however this often results in life-threatening side effects including nephrotoxicity and an increased incidence of cancer and opportunistic infections. Tregs can control graft-vs.-host disease and transplant rejection in experimental models, providing impetus for the use of Tregs as a cellular therapy in clinical transplantation. One of the major barriers to the widespread use of Treg cellular therapy is the requirement to expand cells ex vivo to large numbers in order to alter the overall balance between regulatory and effector cells. Methods that enhance suppressive capacity thereby reducing the need for expansion are therefore of interest. Here, we have compared the function of freshly-isolated and ex vivo-manipulated human Tregs in a pre-clinical humanized mouse model of skin transplantation. Sorted human CD127loCD25+CD4+ Tregs were assessed in three different conditions: freshly-isolated, following transient in vitro activation with antiCD3/antiCD28 beads or after ex vivo-expansion for 2 weeks in the presence of antiCD3/antiCD28 beads and recombinant human IL2. While ex vivo-expansion of human Tregs increased their suppressive function moderately, transient in vitro-activation of freshly isolated Tregs resulted in a powerful enhancement of Treg activity sufficient to promote long-term graft survival of all transplants in vivo. In order to investigate the mechanisms responsible for these effects, we measured the expression of Treg-associated markers and susceptibility to apoptosis in activated Tregs. Transiently activated Tregs displayed enhanced survival and proliferation in vitro and in vivo. On a molecular level, Treg activation resulted in an increased expression of anti-apoptotic BCL2L1 (encoding BCL-XL) which may be at least partially responsible for the observed enhancement in function. Our results suggest that in vitro activation of human Tregs arms them with superior proliferative and survival abilities, enabling them to more effectively control alloresponses. Importantly, this transient activation results in a rapid functional enhancement of freshly-isolated Tregs, thereby providing an opportunity to eliminate the need for in vitro expansion in select circumstances. A protocol employing this technique would therefore benefit from a reduced requirement for large cell numbers for effective therapy.


Subject(s)
Gene Expression Regulation , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , bcl-X Protein/genetics , Animals , Apoptosis/genetics , Apoptosis/immunology , Biomarkers , Cell Survival/genetics , Cell Survival/immunology , Humans , Immune Tolerance , Immunomodulation , Immunophenotyping , Mice , Mice, Knockout , Skin Transplantation , bcl-X Protein/metabolism
6.
J Immunother Cancer ; 7(1): 276, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31651363

ABSTRACT

BACKGROUND: Gene-modified autologous T cells expressing NY-ESO-1c259, an affinity-enhanced T-cell receptor (TCR) reactive against the NY-ESO-1-specific HLA-A*02-restricted peptide SLLMWITQC (NY-ESO-1 SPEAR T-cells; GSK 794), have demonstrated clinical activity in patients with advanced synovial sarcoma (SS). The factors contributing to gene-modified T-cell expansion and the changes within the tumor microenvironment (TME) following T-cell infusion remain unclear. These studies address the immunological mechanisms of response and resistance in patients with SS treated with NY-ESO-1 SPEAR T-cells. METHODS: Four cohorts were included to evaluate antigen expression and preconditioning on efficacy. Clinical responses were assessed by RECIST v1.1. Engineered T-cell persistence was determined by qPCR. Serum cytokines were evaluated by immunoassay. Transcriptomic analyses and immunohistochemistry were performed on tumor biopsies from patients before and after T-cell infusion. Gene-modified T-cells were detected within the TME via an RNAish assay. RESULTS: Responses across cohorts were affected by preconditioning and intra-tumoral NY-ESO-1 expression. Of the 42 patients reported (data cut-off 4June2018), 1 patient had a complete response, 14 patients had partial responses, 24 patients had stable disease, and 3 patients had progressive disease. The magnitude of gene-modified T-cell expansion shortly after infusion was associated with response in patients with high intra-tumoral NY-ESO-1 expression. Patients receiving a fludarabine-containing conditioning regimen experienced increases in serum IL-7 and IL-15. Prior to infusion, the TME exhibited minimal leukocyte infiltration; CD163+ tumor-associated macrophages (TAMs) were the dominant population. Modest increases in intra-tumoral leukocytes (≤5%) were observed in a subset of subjects at approximately 8 weeks. Beyond 8 weeks post infusion, the TME was minimally infiltrated with a TAM-dominant leukocyte infiltrate. Tumor-associated antigens and antigen presentation did not significantly change within the tumor post-T-cell infusion. Finally, NY-ESO-1 SPEAR T cells trafficked to the TME and maintained cytotoxicity in a subset of patients. CONCLUSIONS: Our studies elucidate some factors that underpin response and resistance to NY-ESO-1 SPEAR T-cell therapy. From these data, we conclude that a lymphodepletion regimen containing high doses of fludarabine and cyclophosphamide is necessary for SPEAR T-cell persistence and efficacy. Furthermore, these data demonstrate that non-T-cell inflamed tumors, which are resistant to PD-1/PD-L1 inhibitors, can be treated with adoptive T-cell based immunotherapy. TRIAL REGISTRATION: ClinicalTrials.gov, NCT01343043 , Registered 27 April 2011.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy, Adoptive , Membrane Proteins/immunology , Sarcoma, Synovial/immunology , Sarcoma, Synovial/therapy , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Biomarkers , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Cytokines/metabolism , Cytotoxicity, Immunologic , HLA-A Antigens/immunology , Humans , Immunohistochemistry , Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Sarcoma, Synovial/pathology , T-Cell Antigen Receptor Specificity , Treatment Outcome , Tumor Microenvironment/immunology
7.
Cancer Discov ; 8(8): 944-957, 2018 08.
Article in English | MEDLINE | ID: mdl-29891538

ABSTRACT

We evaluated the safety and activity of autologous T cells expressing NY-ESO-1c259, an affinity-enhanced T-cell receptor (TCR) recognizing an HLA-A2-restricted NY-ESO-1/LAGE1a-derived peptide, in patients with metastatic synovial sarcoma (NY-ESO-1c259T cells). Confirmed antitumor responses occurred in 50% of patients (6/12) and were characterized by tumor shrinkage over several months. Circulating NY-ESO-1c259T cells were present postinfusion in all patients and persisted for at least 6 months in all responders. Most of the infused NY-ESO-1c259T cells exhibited an effector memory phenotype following ex vivo expansion, but the persisting pools comprised largely central memory and stem-cell memory subsets, which remained polyfunctional and showed no evidence of T-cell exhaustion despite persistent tumor burdens. Next-generation sequencing of endogenous TCRs in CD8+ NY-ESO-1c259T cells revealed clonal diversity without contraction over time. These data suggest that regenerative pools of NY-ESO-1c259T cells produced a continuing supply of effector cells to mediate sustained, clinically meaningful antitumor effects.Significance: Metastatic synovial sarcoma is incurable with standard therapy. We employed engineered T cells targeting NY-ESO-1, and the data suggest that robust, self-regenerating pools of CD8+ NY-ESO-1c259T cells produce a continuing supply of effector cells over several months that mediate clinically meaningful antitumor effects despite prolonged exposure to antigen. Cancer Discov; 8(8); 944-57. ©2018 AACR.See related commentary by Keung and Tawbi, p. 914This article is highlighted in the In This Issue feature, p. 899.


Subject(s)
Antigens, Neoplasm/immunology , Membrane Proteins/immunology , Receptors, Antigen, T-Cell/metabolism , Sarcoma, Synovial/therapy , T-Lymphocytes/transplantation , Adoptive Transfer , Adult , CD8-Positive T-Lymphocytes/metabolism , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Pilot Projects , Sarcoma, Synovial/immunology , T-Lymphocytes/immunology , Treatment Outcome , Young Adult
8.
Transplantation ; 101(2): 302-309, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28118317

ABSTRACT

BACKGROUND: Adoptive transfer of forkhead box protein (FOX)3 regulatory T (Treg) cells offers a promising strategy to reduce damage to an allograft by the recipient's immune system. Identification of cell surface markers sufficient to purify Treg cells expanded ex vivo to remove cellular contaminants requires optimization. Furthermore, the expanded Treg must be able to survive, expand, and suppress in allograft recipients exposed to immunosuppressants, such as tacrolimus (TAC). Reduced CD127 expression enhances identification of Treg in the human CD4CD25 population. CD45RA expression identifies naive CD4CD25 Treg with an enhanced stability of Treg phenotype. METHODS: We combine an analysis of CD45RA, CD25, and CD127 expression to identify subpopulations of CD4CD127CD25 cells. Regulatory T cells were sorted according to expression of CD25 and CD45RA and expanded in the presence of a physiological relevant concentration of TAC. Regulatory T cell-specific demethylation region (TSDR) demethylation, FOXP3 expression, and suppression were analyzed. RESULTS: CD4CD127CD25CD45RA Treg cells had a stable TSDR demethylated FOXP3 phenotype after expansion whereas CD4CD127CD25CD45RA Treg cell lost the TSDR demethylated phenotype. CD45RA Treg had a greater capacity to suppress after expansion with TAC. CONCLUSIONS: Although CD45RA Treg retained a greater suppressive capacity when expanded with TAC, the marked loss of the TSDR demethylated status highlights the potential for loss of stability of these cells in transplant recipients treated with TAC based immunosuppression. We show that a population of CD4CD127CD45RA Regulatory T cell may offer the best compromise between susceptibility to loss of suppression when exposed to TAC and maintenance of a TSDR demethylated phenotype following in vitro expansion.


Subject(s)
Calcineurin Inhibitors/pharmacology , DNA Methylation , Immunosuppressive Agents/pharmacology , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-7 Receptor alpha Subunit/immunology , Leukocyte Common Antigens/immunology , T-Lymphocytes, Regulatory/drug effects , Tacrolimus/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Flow Cytometry , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Humans , Immunophenotyping/methods , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Leukocyte Common Antigens/metabolism , Lymphocyte Activation/drug effects , Phenotype , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
9.
Adv Drug Deliv Rev ; 58(8): 948-61, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-17070961

ABSTRACT

Naturally occurring regulatory T cells (Tregs) have been shown to suppress immune responses to self-antigens, thereby limiting autoimmunity. In the case of tumours, where immune responses to self-antigens are beneficial and lead to elimination of the tumour, such suppressive activity is actually detrimental to the host. Manipulation of Tregs holds great promise for the immunotherapy of cancer. Several studies performed using rodent models and indicate that Tregs cells inhibit effective anti-tumour immune responses and that their removal promotes tumour rejection. The increasing number of studies of Tregs in patients with cancer also point to a role for these cells in promoting disease progression. This review summarises the findings of these studies and addresses the advantages and potential pitfalls of manipulating Treg activity for the treatment of cancer.


Subject(s)
Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Immunotherapy , Neoplasms/therapy
10.
J Natl Cancer Inst ; 107(4)2015 Apr.
Article in English | MEDLINE | ID: mdl-25669203

ABSTRACT

Current dogma suggests that tumor-reactive IFN-γ-producing (TH1-type) T-cells are beneficial to patient outcome; however, the clinical consequence of these responses with respect to long-term prognosis in colorectal cancer (CRC) is not understood. Here, we compared the utility of preoperative, peripheral blood-derived IFN-γ(+) T-cell responses specific to carcinoembryonic antigen (CEA), 5T4, or control antigens (n = 64) with tumor staging and clinical details (n = 87) in predicting five-year outcome of CRC patients who underwent resection with curative intent. Although disease recurrence was more likely in patients with stage III tumors, the presence of preoperative, CEA-specific IFN-γ-producing T-cells identified patients at a statistically significantly greater risk of tumor recurrence following surgical resection, irrespective of tumor stage (odds ratio = 5.00, 95% confidence interval = 1.96 to 12.77, two-sided P <.001). Responses to other antigens, including 5T4, did not reflect outcome. Whilst these results initially appear surprising, they could improve prognostication and help redirect adjuvant treatments.


Subject(s)
Biomarkers, Tumor/blood , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/surgery , Interferon-gamma/immunology , Membrane Glycoproteins/immunology , Neoplasm Recurrence, Local/immunology , T-Lymphocytes/immunology , Adult , Aged , Colorectal Neoplasms/pathology , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Odds Ratio , Predictive Value of Tests , Prognosis , Risk Assessment , Risk Factors
11.
PLoS One ; 8(10): e78312, 2013.
Article in English | MEDLINE | ID: mdl-24194918

ABSTRACT

Interleukin (IL)-17 is emerging as an important cytokine in vaccine-induced protection against tuberculosis disease in animal models. Here we show that compared to parenteral delivery, BCG delivered mucosally enhances cytokine production, including interferon gamma and IL-17, in the lungs. Furthermore, we find that cholera toxin, delivered mucosally along with BCG, further enhances IL-17 production by CD4(+) T cells over mucosal BCG alone both in the lungs and systemically. This boosting effect of CT is also observed using a vaccine regimen of BCG followed by the candidate vaccine MVA85A. Using a murine Mycobacterium tuberculosis (M.tb) aerosol challenge model, we demonstrate the ability of cholera toxin delivered at the time of a priming BCG vaccination to improve protection against tuberculosis disease in a manner at least partially dependent on the observed increase in IL-17. This observed increase in IL-17 in the lungs has no adverse effect on lung pathology following M.tb challenge, indicating that IL-17 can safely be boosted in murine lungs in a vaccine/M.tb challenge setting.


Subject(s)
BCG Vaccine/pharmacology , Cholera Toxin/pharmacology , Interleukin-17/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/prevention & control , Analysis of Variance , Animals , BCG Vaccine/immunology , Mice , Statistics, Nonparametric , Tuberculosis/immunology
12.
PLoS One ; 7(12): e50447, 2012.
Article in English | MEDLINE | ID: mdl-23284637

ABSTRACT

The Bacillus Calmette - Guerin (BCG) vaccine provides a critical but limited defense against Mycobacterium tuberculosis (M.tb). More than 60 years after the widespread introduction of BCG, there is an urgent need for a better vaccine. A large body of pre-clinical research continues to support ongoing clinical trials to assess whether viral vectors expressing M.tb antigens that are shared by BCG and M.tb, can be used alongside BCG to enhance protection. A major focus involves using multiple unique viral vectors to limit anti-vector immunity and thereby enhance responses to the insert antigen delivered. The successful introduction of viral vector vaccines to target M.tb and other pathogens will be reliant on reducing the costs when using multiple vectors and inhibiting the development of unwanted anti-vector responses that interfere with the response to insert antigen. This study examines methods to reduce the logistical costs of vaccination by mixing different viral vectors that share the same insert antigen in one vaccine; and whether combining different viral vectors reduces anti-vector immunity to improve immunogenicity to the insert antigen. Here we show that a homologous prime-boost regimen with a mixture of MVA (Modified Vaccinia virus Ankara) and Ad5 (human adenovirus type 5) vectors both expressing Ag85A in a single vaccine preparation is able to reduce anti-vector immunity, compared with a homologous prime-boost regimen with either vector alone. However, the level of immunogenicity induced by the homologous mixture remained comparable to that induced with single viral vectors and was less immunogenic than a heterologous Ad5 prime-MVA-boost regimen. These findings advance the understanding of how anti-vector immunity maybe reduced in viral vector vaccination regimens. Furthermore, an insight is provided to the impact on vaccine immunogenicity from altering vaccination methods to reduce the logistical demands of using separate vaccine preparations in the field.


Subject(s)
Acyltransferases/genetics , Acyltransferases/immunology , Adenoviridae/genetics , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Genetic Vectors/genetics , Vaccination/methods , Vaccinia virus/genetics , Amino Acid Sequence , Animals , Bacterial Vaccines/genetics , Female , Gene Expression , Immunization, Secondary , Injections , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mycobacterium tuberculosis/immunology
13.
Cancer Res ; 71(3): 736-46, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21156649

ABSTRACT

A significant enrichment of CD4(+)Foxp3(+) T cells (regulatory T cells, Treg) is frequently observed in murine and human carcinomas. As Tregs can limit effective antitumor immune responses, thereby promoting tumor progression, it is important that the mechanisms underpinning intratumoral accumulation of Tregs are identified. Because of evidence gathered mostly in vitro, the conversion of conventional T cells (Tconv) into Tregs has been proposed as one such mechanism. We assessed the contribution of conversion in vivo by analyzing the TCR (T-cell receptor) repertoires of Tconvs and Tregs in carcinogen-induced tumors in mice. Our results indicate that the TCR repertoires of Tregs and Tconvs within tumor-infiltrating lymphocytes (TIL) are largely distinct. Indeed, the cell population with the greatest degree of repertoire similarity with tumor-infiltrating Tregs was the Treg population from the tumor-draining lymph node. These findings demonstrate that conversion of Tconvs does not contribute significantly to the accumulation of tumor-infiltrating Tregs; rather, Tconvs and Tregs arise from different populations with unique TCR repertoires. Enrichment of Tregs within TILs most likely, therefore, reflects differences in the way that Tregs and Tconvs are influenced by the tumor microenvironment. Elucidating the nature of these influences may indicate how the balance between tumor-infiltrating Tregs and Tconvs can be manipulated for therapeutic purposes.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Experimental/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Carcinogens , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Methylcholanthrene , Mice , Mice, Transgenic , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics
15.
Eur J Immunol ; 37(3): 758-67, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17294404

ABSTRACT

CD4+CD25+ regulatory T cells (Treg) are known to influence T cell responses to tumours. Here we have explored the role of Treg in inhibiting not only adaptive, but also innate immune responses to tumours. To this end we used a Fas ligand (FasL)-expressing melanoma cell line in a mouse model. In this system, innate immunity is sufficient to reject the tumour. All mice depleted of Treg and challenged with FasL-expressing melanoma remained tumour-free. Investigation of the underlying cellular effector mechanisms revealed that depletion of Treg enhanced an NK cell response capable of tumour lysis. Furthermore, this initial innate immune response primed mice to make an effective adaptive immune response leading to complete rejection of challenge with the parental melanoma. Both antigen-specific antibody and CD4+ T cells were implicated in protection via adaptive immunity. We conclude that removal of Treg and vaccination with whole tumour cells expressing FasL activates multiple arms of the immune system, leading to efficient tumour rejection. These findings highlight a novel role for FasL in inducing innate immune responses that are normally inhibited by Treg and uncover an adjuvant effect of FasL that can be used to stimulate tumour immunity after depletion of Treg.


Subject(s)
Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/physiology , Melanoma, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Cells, Cultured , Immunity, Active , Immunity, Innate , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
16.
PLoS One ; 1: e129, 2006 Dec 27.
Article in English | MEDLINE | ID: mdl-17205133

ABSTRACT

BACKGROUND: A wealth of evidence obtained using mouse models indicates that CD4(+)CD25(+)FOXP3(+) regulatory T cells (Treg) maintain peripheral tolerance to self-antigens and also inhibit anti-tumor immune responses. To date there is limited information about CD4(+) T cell responses in patients with colorectal cancer (CRC). We set out to measure T cell responses to a tumor-associated antigen and examine whether Treg impinge on those anti-tumor immune responses in CRC patients. METHODOLOGY AND PRINCIPAL FINDINGS: Treg were identified and characterized as CD4(+)CD25(+)FOXP3(+) using flow cytometry. An increased frequency of Treg was demonstrated in both peripheral blood and mesenteric lymph nodes of patients with colorectal cancer (CRC) compared with either healthy controls or patients with inflammatory bowel disease (IBD). Depletion of Treg from peripheral blood mononuclear cells (PBMC) of CRC patients unmasked CD4(+) T cell responses, as observed by IFNgamma release, to the tumor associated antigen 5T4, whereas no effect was observed in a healthy age-matched control group. CONCLUSIONS/SIGNIFICANCE: Collectively, these data demonstrate that Treg capable of inhibiting tumor associated antigen-specific immune responses are enriched in patients with CRC. These results support a rationale for manipulating Treg to enhance cancer immunotherapy.


Subject(s)
Colorectal Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Adenocarcinoma/immunology , Antigens, Neoplasm , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Case-Control Studies , Forkhead Transcription Factors/metabolism , Humans , In Vitro Techniques , Interferon-gamma/biosynthesis , Interleukin-2 Receptor alpha Subunit/metabolism , Lymph Nodes/immunology , Lymphocyte Activation , Lymphocyte Depletion , Membrane Glycoproteins/immunology , Self Tolerance
SELECTION OF CITATIONS
SEARCH DETAIL