Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 13(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830955

ABSTRACT

Endometrial cancer (EC) is a common gynecological malignancy and the fourth most common malignancy in European and North American women. Amongst EC, the advanced serous, p53-mutated, and pMMR subtypes have the highest risk of relapse despite optimal standard of care therapy. At present, there is no standard of care maintenance treatment to prevent relapse among these high-risk patients. Vaccines are a form of immunotherapy that can potentially increase the immunogenicity of pMMR, serous, and p53-mutated tumors to render them responsive to check point inhibitor-based immunotherapy. We demonstrate, for the first time, the feasibility of generating a personalized dendritic cell vaccine pulsed with peptide neoantigens in a patient with pMMR, p53-mutated, and serous endometrial adenocarcinoma (SEC). The personalized vaccine was administered in combination with systemic chemotherapy to treat an inoperable metastatic recurrence. This treatment association demonstrated the safety and immunogenicity of the personalized dendritic cell vaccine. Interestingly, a complete oncological response was obtained with respect to both radiological assessment and the tumor marker CA-125.

2.
Cytotherapy ; 22(12): 780-791, 2020 12.
Article in English | MEDLINE | ID: mdl-33069566

ABSTRACT

BACKGROUND AIMS: Several studies report on Good Manufacturing Process (GMP)-compliant manufacturing protocols for the ex vivo expansion of tumor-infiltrating lymphocytes (TILs) for the treatment of patients with refractory melanoma and other solid malignancies. Further opportunities for improvements in terms of ergonomy and operating time have been identified. METHODS: To enable GMP-compliant TILs production for adoptive cell therapy needs, a simple automated and reproducible protocol for TILs manufacturing with the use of a closed system was developed and implemented at the authors' institution. RESULTS: This protocol enabled significant operating time reduction during TILs expansion while allowing the generation of high-quality TILs products. CONCLUSIONS: A simplified and efficient method of TILs expansion will enable the broadening of individualized tumor therapy and will increase patients' access to state-of-the-art TILs adoptive cell therapy treatment.


Subject(s)
Cell Culture Techniques/methods , Hospitals , Lymphocytes, Tumor-Infiltrating/cytology , Automation , Cell Count , Cell Proliferation , Cryopreservation , Female , Humans , Kinetics , Phenotype , Quality Control
3.
Curr Opin Biotechnol ; 65: 190-196, 2020 10.
Article in English | MEDLINE | ID: mdl-32334152

ABSTRACT

New treatments are urgently needed in patients with ovarian cancer (OC), as diagnosis is delayed in many instances, resulting in 85% recurrence of the disease following surgery and standard chemotherapy. OC is considered to be an immunological type of cancer, despite its limited response to current immunotherapy options, including vaccination. Thus, additional interventions may improve their efficacy. Dendritic cells (DCs) are the most widely used cellular vaccination therapy in patients with OC due to their crucial role in the initiation and development of immune response. There are viable options for DC-vaccination with a favorable toxicity profile, but specific alternatives should consider the limited therapeutic effectiveness of DC-vaccination in OC treatment. In this respect, B-cells and macrophages provide additional possibilities that may be explored for immunotherapy. Here we consider the current state-of-the-art of immunotherapy strategies for OC treatment and evaluate their potential for future improvements.


Subject(s)
Cancer Vaccines , Ovarian Neoplasms , Vaccines , Cancer Vaccines/therapeutic use , Dendritic Cells , Female , Humans , Immunotherapy , Ovarian Neoplasms/therapy
4.
Vaccines (Basel) ; 8(1)2020 Jan 14.
Article in English | MEDLINE | ID: mdl-31947581

ABSTRACT

With the emergence of immune checkpoint inhibitors and adoptive T-cell therapies, there is a considerable interest in using personalized autologous dendritic cell (DC) vaccines in combination with T cell-targeting immunotherapies to potentially maximize the therapeutic impact of DC vaccines. Here, we describe the development and optimization of a Good Manufacturing Practice (GMP)-compliant manufacturing process based on tumor lysate as a tumor antigen source for the production of an oxidized tumor cell lysate loaded DC (OC-DC) vaccine. The manufacturing process required one day for lysate preparation and six days for OC-DC vaccine production. Tumor lysate production was standardized based on an optimal tumor digestion protocol and the immunogenicity was improved through oxidation using hypochloric acid prior to freeze-thaw cycles resulting in the oxidized tumor cell lysate (OC-L). Next, monocytes were selected using the CliniMACS prodigy closed system and were placed in culture in cell factories in the presence of IL-4 and GM-CSF. Immature DCs were loaded with OC-L and matured using MPLA-IFNγ. After assessing the functionality of the OC-DC cells (IL12p70 secretion and COSTIM assay), the OC-DC vaccine was cryopreserved in multiple doses for single use. Finally, the stability of the formulated doses was tested and validated. We believe this GMP-compliant DC vaccine manufacturing process will facilitate access of patients to personalized DC vaccines, and allow for multi-center clinical trials.

5.
J Transl Med ; 17(1): 391, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31771601

ABSTRACT

BACKGROUND: Most ovarian cancer patients are diagnosed at a late stage with 85% of them relapsing after surgery and standard chemotherapy; for this reason, new treatments are urgently needed. Ovarian cancer has become a candidate for immunotherapy by reason of their expression of shared tumor-associated antigens (TAAs) and private mutated neoantigens (NeoAgs) and the recognition of the tumor by the immune system. Additionally, the presence of intraepithelial tumor infiltrating lymphocytes (TILs) is associated with improved progression-free and overall survival of patients with ovarian cancer. The aim of active immunotherapy, including vaccination, is to generate a new anti-tumor response and amplify an existing immune response. Recently developed NeoAgs-based cancer vaccines have the advantage of being more tumor specific, reducing the potential for immunological tolerance, and inducing robust immunogenicity. METHODS: We propose a randomized phase I/II study in patients with advanced ovarian cancer to compare the immunogenicity and to assess safety and feasibility of two personalized DC vaccines. After standard of care surgery and chemotherapy, patients will receive either a novel vaccine consisting of autologous DCs pulsed with up to ten peptides (PEP-DC), selected using an agnostic, yet personalized, epitope discovery algorithm, or a sequential combination of a DC vaccine loaded with autologous oxidized tumor lysate (OC-DC) prior to an equivalent PEP-DC vaccine. All vaccines will be administered in combination with low-dose cyclophosphamide. This study is the first attempt to compare the two approaches and to use NeoAgs-based vaccines in ovarian cancer in the adjuvant setting. DISCUSSION: The proposed treatment takes advantage of the beneficial effects of pre-treatment with OC-DC prior to PEP-DC vaccination, prompting immune response induction against a wide range of patient-specific antigens, and amplification of pre-existing NeoAgs-specific T cell clones. Trial registration This trial is already approved by Swissmedic (Ref.: 2019TpP1004) and will be registered at http://www.clinicaltrials.gov before enrollment opens.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Peptides/immunology , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Female , Humans , Neoplasm Grading , Neoplasm Staging , Transplantation, Autologous
7.
Front Immunol ; 10: 1832, 2019.
Article in English | MEDLINE | ID: mdl-31440238

ABSTRACT

Despite the promising therapeutic effects of immune checkpoint blockade (ICB), most patients with solid tumors treated with anti-PD-1/PD-L1 monotherapy do not achieve objective responses, with most tumor regressions being partial rather than complete. It is hypothesized that the absence of pre-existing antitumor immunity and/or the presence of additional tumor immune suppressive factors at the tumor microenvironment are responsible for such therapeutic failures. It is therefore clear that in order to fully exploit the potential of PD-1 blockade therapy, antitumor immune response should be amplified, while tumor immune suppression should be further attenuated. Cancer vaccines may prime patients for treatments with ICB by inducing effective anti-tumor immunity, especially in patients lacking tumor-infiltrating T-cells. These "non-inflamed" non-permissive tumors that are resistant to ICB could be rendered sensitive and transformed into "inflamed" tumor by vaccination. In this article we describe a clinical study where we use pancreatic cancer as a model, and we hypothesize that effective vaccination in pancreatic cancer patients, along with interventions that can reprogram important immunosuppressive factors in the tumor microenvironment, can enhance tumor immune recognition, thus enhancing response to PD-1/PD-L1 blockade. We incorporate into the schedule of standard of care (SOC) chemotherapy adjuvant setting a vaccine platform comprised of autologous dendritic cells loaded with personalized neoantigen peptides (PEP-DC) identified through our own proteo-genomics antigen discovery pipeline. Furthermore, we add nivolumab, an antibody against PD-1, to boost and maintain the vaccine's effect. We also demonstrate the feasibility of identifying personalized neoantigens in three pancreatic ductal adenocarcinoma (PDAC) patients, and we describe their optimal incorporation into long peptides for manufacturing into vaccine products. We finally discuss the advantages as well as the scientific and logistic challenges of such an exploratory vaccine clinical trial, and we highlight its novelty.


Subject(s)
Antigens, Neoplasm/administration & dosage , Antineoplastic Agents, Immunological/therapeutic use , Aspirin/therapeutic use , Carcinoma, Pancreatic Ductal/therapy , Chemotherapy, Adjuvant , Dendritic Cells/immunology , Immunotherapy, Active/methods , Molecular Targeted Therapy , Nivolumab/therapeutic use , Pancreatic Neoplasms/therapy , Amino Acid Sequence , Carcinoma, Pancreatic Ductal/drug therapy , Combined Modality Therapy , Exome , Feasibility Studies , Humans , Neoplasm Proteins/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Peptides/immunology , Precision Medicine , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Proof of Concept Study , Sequence Alignment , Sequence Homology, Nucleic Acid
8.
Front Immunol ; 10: 766, 2019.
Article in English | MEDLINE | ID: mdl-31031762

ABSTRACT

With the advent of combined immunotherapies, personalized dendritic cell (DC)-based vaccination could integrate the current standard of care for the treatment of a large variety of tumors. Due to their proficiency at antigen presentation, DC are key coordinators of the innate and adaptive immune system, and have critical roles in the induction of antitumor immunity. However, despite proven immunogenicity and favorable safety profiles, DC-based immunotherapies have not succeeded at inducing significant objective clinical responses. Emerging data suggest that the combination of DC-based vaccination with other cancer therapies may fully unleash the potential of DC-based cancer vaccines and improve patient survival. In this review, we discuss the recent efforts to develop innovative personalized DC-based vaccines and their use in combined therapies, with a particular focus on ovarian cancer and the promising results of mutanome-based personalized immunotherapies.


Subject(s)
Cancer Vaccines , Dendritic Cells , Precision Medicine , Dendritic Cells/immunology , Dendritic Cells/transplantation , Humans , Immunotherapy/methods
9.
Immunology ; 152(3): 425-438, 2017 11.
Article in English | MEDLINE | ID: mdl-28640942

ABSTRACT

The success of immune system-based cancer therapies depends on a broad immune response engaging a range of effector cells and mechanisms. Immune mobilizing monoclonal T cell receptors (TCRs) against cancer (ImmTAC™ molecules: fusion proteins consisting of a soluble, affinity enhanced TCR and an anti-CD3 scFv antibody) were previously shown to redirect CD8+ and CD4+ T cells against tumours. Here we present evidence that IMCgp100 (ImmTAC recognizing a peptide derived from the melanoma-specific protein, gp100, presented by HLA-A*0201) efficiently redirects and activates effector and memory cells from both CD8+ and CD4+ repertoires. Using isolated subpopulations of T cells, we find that both terminally differentiated and effector memory CD8+ T cells redirected by IMCgp100 are potent killers of melanoma cells. Furthermore, CD4+ effector memory T cells elicit potent cytotoxic activity leading to melanoma cell killing upon redirection by IMCgp100. The majority of T cell subsets belonging to both the CD8+ and CD4+ repertoires secrete key pro-inflammatory cytokines (tumour necrosis factor-α, interferon-γ, interleukin-6) and chemokines (macrophage inflammatory protein-1α-ß, interferon-γ-inducible protein-10, monocyte chemoattractant protein-1). At an individual cell level, IMCgp100-redirected T cells display a polyfunctional phenotype, which is a hallmark of a potent anti-cancer response. This study demonstrates that IMCgp100 induces broad immune responses that extend beyond the induction of CD8+ T cell-mediated cytotoxicity. These findings are of particular importance because IMCgp100 is currently undergoing clinical trials as a single agent or in combination with check point inhibitors for patients with malignant melanoma.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Melanoma/therapy , Proteins/pharmacology , Single-Chain Antibodies/pharmacology , Skin Neoplasms/therapy , gp100 Melanoma Antigen/immunology , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Dose-Response Relationship, Drug , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , Immunologic Memory/drug effects , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Phenotype , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Time Factors , gp100 Melanoma Antigen/metabolism
10.
J Immunol ; 193(6): 2784-91, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25127860

ABSTRACT

Protection against reinfection is mediated by Ag-specific memory CD8 T cells, which display stem cell-like function. Because canonical Wnt (Wingless/Int1) signals critically regulate renewal versus differentiation of adult stem cells, we evaluated Wnt signal transduction in CD8 T cells during an immune response to acute infection with lymphocytic choriomeningitis virus. Whereas naive CD8 T cells efficiently transduced Wnt signals, at the peak of the primary response to infection only a fraction of effector T cells retained signal transduction and the majority displayed strongly reduced Wnt activity. Reduced Wnt signaling was in part due to the downregulation of Tcf-1, one of the nuclear effectors of the pathway, and coincided with progress toward terminal differentiation. However, the correlation between low and high Wnt levels with short-lived and memory precursor effector cells, respectively, was incomplete. Adoptive transfer studies showed that low and high Wnt signaling did not influence cell survival but that Wnt high effectors yielded memory cells with enhanced proliferative potential and stronger protective capacity. Likewise, following adoptive transfer and rechallenge, memory cells with high Wnt levels displayed increased recall expansion, compared with memory cells with low Wnt signaling, which were preferentially effector-like memory cells, including tissue-resident memory cells. Thus, canonical Wnt signaling identifies CD8 T cells with enhanced proliferative potential in part independent of commonly used cell surface markers to discriminate effector and memory T cell subpopulations. Interventions that maintain Wnt signaling may thus improve the formation of functional CD8 T cell memory during vaccination.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Wnt Proteins/immunology , Wnt Signaling Pathway/immunology , Adoptive Transfer , Animals , Axin Protein/biosynthesis , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Cell Proliferation , Down-Regulation , Hepatocyte Nuclear Factor 1-alpha/biosynthesis , Immunologic Memory/immunology , Lectins, C-Type , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Immunologic/biosynthesis , T-Lymphocyte Subsets/immunology , Vaccination
12.
Clin Transl Allergy ; 3(1): 17, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23725004

ABSTRACT

BACKGROUND: Synthetic contiguous overlapping peptides (COPs) may represent an alternative to allergen extracts or recombinant allergens for allergen specific immunotherapy. In combination, COPs encompass the entire allergen sequence, providing all potential T cell epitopes, while preventing IgE conformational epitopes of the native allergen. METHODS: Individual COPs were derived from the sequence of Bet v 1, the major allergen of birch pollen, and its known crystal structure, and designed to avoid IgE binding. Three sets of COPs were tested in vitro in competition ELISA and basophil degranulation assays. Their in vivo reactivity was determined by intraperitoneal challenge in rBet v 1 sensitized mice as well as by skin prick tests in volunteers with allergic rhinoconjunctivitis to birch pollen. RESULTS: The combination, named AllerT, of three COPs selected for undetectable IgE binding in competition assays and for the absence of basophil activation in vitro was unable to induce anaphylaxis in sensitized mice in contrast to rBet v 1. In addition no positive reactivity to AllerT was observed in skin prick tests in human volunteers allergic to birch pollen. In contrast, a second set of COPs, AllerT4-T5 displayed some residual IgE binding in competition ELISA and a weak subliminal reactivity to skin prick testing. CONCLUSIONS: The hypoallergenicity of contiguous overlapping peptides was confirmed by low, if any, IgE binding activity in vitro, by the absence of basophil activation and the absence of in vivo induction of allergic reactions in mouse and human. TRIAL REGISTRATION: ClinicalTrials.gov NCT01719133.

13.
Immunity ; 38(4): 742-53, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601686

ABSTRACT

MicroRNAs (miRNAs) regulate the function of several immune cells, but their role in promoting CD8(+) T cell immunity remains unknown. Here we report that miRNA-155 is required for CD8(+) T cell responses to both virus and cancer. In the absence of miRNA-155, accumulation of effector CD8(+) T cells was severely reduced during acute and chronic viral infections and control of virus replication was impaired. Similarly, Mir155(-/-) CD8(+) T cells were ineffective at controlling tumor growth, whereas miRNA-155 overexpression enhanced the antitumor response. miRNA-155 deficiency resulted in accumulation of suppressor of cytokine signaling-1 (SOCS-1) causing defective cytokine signaling through STAT5. Consistently, enforced expression of SOCS-1 in CD8(+) T cells phenocopied the miRNA-155 deficiency, whereas SOCS-1 silencing augmented tumor destruction. These findings identify miRNA-155 and its target SOCS-1 as key regulators of effector CD8(+) T cells that can be modulated to potentiate immunotherapies for infectious diseases and cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Melanoma, Experimental/immunology , MicroRNAs/metabolism , Adoptive Transfer , Animals , Apoptosis/genetics , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , RNA, Small Interfering/genetics , STAT6 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Virus Replication/genetics
14.
Int J Inflam ; 2012: 686739, 2012.
Article in English | MEDLINE | ID: mdl-22762009

ABSTRACT

Introduction. Preclinical and clinical evidences for a role of oral probiotics in the management of allergic diseases are emerging. Aim. We aimed at testing the immunomodulatory effects of intranasal versus intragastric administration of Lactobacillus paracasei NCC2461 in a mouse model of allergic airway inflammation and the specificity of different probiotics by comparing L. paracasei NCC2461 to Lactobacillus plantarum NCC1107. Methods. L. paracasei NCC2461 or L. plantarum NCC1107 strains were administered either intragastrically (NCC2461) or intranasally (NCC2461 or NCC1107) to OVA-sensitized mice challenged with OVA aerosols. Inflammatory cell recruitment into BALF, eotaxin and IL-5 production in the lungs were measured. Results. Intranasal L. paracasei NCC2461 efficiently protected sensitized mice upon exposure to OVA aerosols in a dose-dependent manner as compared to control mice. Inflammatory cell number, eotaxin and IL-5 were significantly reduced in BALF. Intranasal supplementation of L. paracasei NCC2461 was more potent than intragastric application in limiting the allergic response and possibly linked to an increase in T regulatory cells in the lungs. Finally, intranasal L. plantarum NCC1107 reduced total and eosinophilic lung inflammation, but increased neutrophilia and macrophages infiltration. Conclusion. A concerted selection of intervention schedule, doses, and administration routes (intranasal versus intragastric) may markedly contribute to modulate airway inflammation in a probiotic strain-specific manner.

15.
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
16.
Proc Natl Acad Sci U S A ; 107(21): 9777-82, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20457902

ABSTRACT

Immune protection from intracellular pathogens depends on the generation of terminally differentiated effector and of multipotent memory precursor CD8 T cells, which rapidly regenerate effector and memory cells during recurrent infection. The identification of factors and pathways involved in CD8 T cell differentiation is of obvious importance to improve vaccination strategies. Here, we show that mice lacking T cell factor 1 (Tcf-1), a nuclear effector of the canonical Wingless/Integration 1 (Wnt) signaling pathway, mount normal effector and effector memory CD8 T cell responses to infection with lymphocytic choriomeningitis virus (LCMV). However, Tcf-1-deficient CD8 T cells are selectively impaired in their ability to expand upon secondary challenge and to protect from recurrent virus infection. Tcf-1-deficient mice essentially lack CD8 memory precursor T cells, which is evident already at the peak of the primary response, suggesting that Tcf-1 programs CD8 memory cell fate. The function of Tcf-1 to establish CD8 T cell memory is dependent on the catenin-binding domain in Tcf-1 and requires the Tcf-1 coactivators and Wnt signaling intermediates beta-catenin and gamma-catenin. These findings demonstrate that the canonical Wnt signaling pathway plays an essential role for CD8 central memory T cell differentiation under physiological conditions in vivo. They raise the possibility that modulation of Wnt signaling may be exploited to improve the generation of CD8 memory T cells during vaccination or for therapies designed to promote sustained cytotoxic CD8 T cell responses against tumors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Signal Transduction , T Cell Transcription Factor 1/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Hepatocyte Nuclear Factor 1-alpha , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Protein Binding , T Cell Transcription Factor 1/deficiency , Wnt Proteins/metabolism , beta Catenin/immunology , beta Catenin/metabolism , gamma Catenin/immunology , gamma Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...