Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Ther ; 31(1): 48-65, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36045586

ABSTRACT

Regulatory T cells overwhelm conventional T cells in the tumor microenvironment (TME) thanks to a FOXP3-driven metabolic program that allows them to engage different metabolic pathways. Using a melanoma model of adoptive T cell therapy (ACT), we show that FOXP3 overexpression in mature CD8 T cells improved their antitumor efficacy, favoring their tumor recruitment, proliferation, and cytotoxicity. FOXP3-overexpressing (Foxp3UP) CD8 T cells exhibited features of tissue-resident memory-like and effector T cells, but not suppressor activity. Transcriptomic analysis of tumor-infiltrating Foxp3UP CD8 T cells showed positive enrichment in a wide variety of metabolic pathways, such as glycolysis, fatty acid (FA) metabolism, and oxidative phosphorylation (OXPHOS). Intratumoral Foxp3UP CD8 T cells exhibited an enhanced capacity for glucose and FA uptake as well as accumulation of intracellular lipids. Interestingly, Foxp3UP CD8 T cells compensated for the loss of mitochondrial respiration-driven ATP production by activating aerobic glycolysis. Moreover, in limiting nutrient conditions these cells engaged FA oxidation to drive OXPHOS for their energy demands. Importantly, their ability to couple glycolysis and OXPHOS allowed them to sustain proliferation under glucose restriction. Our findings demonstrate a hitherto unknown role for FOXP3 in the adaptation of CD8 T cells to TME that may enhance their efficacy in ACT.


Subject(s)
CD8-Positive T-Lymphocytes , Forkhead Transcription Factors , Immunotherapy, Adoptive , Melanoma , Humans , CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Glucose/metabolism , Melanoma/therapy , Tumor Microenvironment
2.
J Immunother Cancer ; 9(11)2021 11.
Article in English | MEDLINE | ID: mdl-34810235

ABSTRACT

BACKGROUND: Target antigen (Ag) loss has emerged as a major cause of relapse after chimeric antigen receptor T (CART)-cell therapy. We reasoned that the combination of CART cells, with the consequent tumor debulking and release of Ags, together with an immunomodulatory agent, such as the stimulator of interferon gene ligand (STING-L) 2'3'-cyclic GMP-AMP (2'3'-cGAMP), may facilitate the activation of an endogenous response to secondary tumor Ags able to counteract this tumor escape mechanism. METHODS: Mice bearing B16-derived tumors expressing prostate-specific membrane Ag or gp75 were treated systemically with cognate CART cells followed by intratumoral injections of 2'3'-cGAMP. We studied the target Ag inmunoediting by CART cells and the effect of the CART/STING-L combination on the control of STING-L-treated and STING-L-non-treated tumors and on the endogenous antitumor T-cell response. The role of Batf3-dependent dendritic cells (DCs), stimulator of interferon gene (STING) signaling and perforin (Perf)-mediated killing in the efficacy of the combination were analyzed. RESULTS: Using an immune-competent solid tumor model, we showed that CART cells led to the emergence of tumor cells that lose the target Ag, recreating the cancer immunoediting effect of CART-cell therapy. In this setting, the CART/STING-L combination, but not the monotherapy with CART cells or STING-L, restrained tumor progression and enhanced overall survival, showing abscopal effects on distal STING-L-non-treated tumors. Interestingly, a secondary immune response against non-chimeric antigen receptor-targeted Ags (epitope spreading), as determined by major histocompatibility complex-I-tetramer staining, was fostered and its intensity correlated with the efficacy of the combination. This was consistent with the oligoclonal expansion of host T cells, as revealed by in-depth T-cell receptor repertoire analysis. Moreover, only in the combination group did the activation of endogenous T cells translate into a systemic antitumor response. Importantly, the epitope spreading and the antitumor effects of the combination were fully dependent on host STING signaling and Batf3-dependent DCs, and were partially dependent on Perf release by CART cells. Interestingly, the efficacy of the CART/STING-L treatment also depended on STING signaling in CART cells. CONCLUSIONS: Our data show that 2'3'-cGAMP is a suitable adjuvant to combine with CART-cell therapy, allowing the induction of an endogenous T-cell response that prevents the outgrowth of Ag-loss tumor variants.


Subject(s)
Epitopes/genetics , Immunotherapy, Adoptive/methods , Immunotherapy/methods , Neoplasms/genetics , Tumor Escape/genetics , Animals , Humans , Male , Mice
3.
Int J Mol Sci ; 22(8)2021 Apr 17.
Article in English | MEDLINE | ID: mdl-33920699

ABSTRACT

Immune checkpoint inhibitors (ICIs) have demonstrated remarkable efficacy in a growing number of malignancies. However, overcoming primary or secondary resistances is difficult due to pharmacokinetics issues and side effects associated with high systemic exposure. Local or regional expression of monoclonal antibodies (mAbs) using gene therapy vectors can alleviate this problem. In this work, we describe a high-capacity adenoviral vector (HCA-EFZP-aPDL1) equipped with a mifepristone-inducible system for the controlled expression of an anti-programmed death ligand 1 (PD-L1) blocking antibody. The vector was tested in an immune-competent mouse model of colorectal cancer based on implantation of MC38 cells. A single local administration of HCA-EFZP-aPDL1 in subcutaneous lesions led to a significant reduction in tumor growth with minimal release of the antibody in the circulation. When the vector was tested in a more stringent setting (rapidly progressing peritoneal carcinomatosis), the antitumor effect was marginal even in combination with other immune-stimulatory agents such as polyinosinic-polycytidylic acid (pI:C), blocking mAbs for T cell immunoglobulin, mucin-domain containing-3 (TIM-3) or agonistic mAbs for 4-1BB (CD137). In contrast, macrophage depletion by clodronate liposomes enhanced the efficacy of HCA-EFZP-aPDL1. These results highlight the importance of addressing macrophage-associated immunoregulatory mechanisms to overcome resistance to ICIs in the context of colorectal cancer.


Subject(s)
Antibodies, Blocking/genetics , B7-H1 Antigen/metabolism , Carcinoma/therapy , Genetic Therapy/methods , Immunotherapy/methods , Macrophages/immunology , Peritoneal Neoplasms/therapy , Adenoviridae/genetics , Animals , Antibodies, Blocking/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Cell Line , Female , Genetic Vectors/genetics , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immunologic Factors/therapeutic use , Mice , Mice, Inbred C57BL , Poly I-C/therapeutic use
4.
Br J Cancer ; 124(6): 1138-1149, 2021 03.
Article in English | MEDLINE | ID: mdl-33402737

ABSTRACT

BACKGROUND: Adoptive immunotherapy with tumour-infiltrating lymphocytes (TIL) may benefit from the use of selective markers, such as PD-1, for tumour-specific T-cell enrichment, and the identification of predictive factors that help identify those patients capable of rendering tumour-reactive TILs. We have investigated this in ovarian cancer (OC) patients as candidates for TIL therapy implementation. METHODS: PD-1- and PD-1+ CD8 TILs were isolated from ovarian tumours and expanded cells were tested against autologous tumour cells. Baseline tumour samples were examined using flow cytometry, multiplexed immunofluorescence and Nanostring technology, for gene expression analyses, as well as a next-generation sequencing gene panel, for tumour mutational burden (TMB) calculation. RESULTS: Tumour-reactive TILs were detected in half of patients and were exclusively present in cells derived from the PD-1+ fraction. Importantly, a high TIL density in the fresh tumour, the presence of CD137+ cells within the PD-1+CD8+ TIL subset and their location in the tumour epithelium, together with a baseline T-cell-inflamed genetic signature and/or a high TMB, are features that identify patients rendering tumour-reactive TIL products. CONCLUSION: We have demonstrated that PD-1 identifies ovarian tumour-specific CD8 TILs and has uncovered predictive factors that identify OC patients who are likely to render tumour-specific cells from PD-1+ TILs.


Subject(s)
Biomarkers, Tumor/genetics , CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation, Neoplastic , Lymphocytes, Tumor-Infiltrating/immunology , Ovarian Neoplasms/pathology , Programmed Cell Death 1 Receptor/metabolism , Biomarkers, Tumor/metabolism , Female , Follow-Up Studies , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Phenotype , Prognosis , Retrospective Studies
5.
Cancer Cell ; 36(6): 613-629.e7, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31761658

ABSTRACT

Retroviral gene transfer of interleukin-12 (IL-12) into T cells markedly enhances antitumor efficacy upon adoptive transfer but has clinically shown unacceptable severe side effects. To overcome the toxicity, we engineered tumor-specific CD8+ T cells to transiently express IL-12. Engineered T cells injected intratumorally, but not intravenously, led to complete rejections not only of the injected lesion but also of distant concomitant tumors. Efficacy was further enhanced by co-injection with agonist anti-CD137 mAb or by transient co-expression of CD137 ligand. This treatment induced epitope spreading of the endogenous CD8+ T cell immune response in a manner dependent on cDC1 dendritic cells. Mouse and human tumor-infiltrating T lymphocyte cultures can be transiently IL-12 engineered to attain marked immunotherapeutic effects.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive , Interleukin-12/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Adoptive Transfer/methods , Animals , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Dendritic Cells/immunology , Humans , Immunotherapy, Adoptive/methods , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Mice , T-Lymphocytes, Cytotoxic/immunology
6.
Mol Ther ; 27(11): 1892-1905, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31563534

ABSTRACT

Immune checkpoint blockade has shown anti-cancer efficacy, but requires systemic administration of monoclonal antibodies (mAbs), often leading to adverse effects. To avoid toxicity, mAbs could be expressed locally in tumors. We developed adeno-associated virus (AAV) and Semliki Forest virus (SFV) vectors expressing anti-programmed death ligand 1 (aPDL1) mAb. When injected intratumorally in MC38 tumors, both viral vectors led to similar local mAb expression at 24 h, diminishing quickly in SFV-aPDL1-treated tumors. However, SFV-aPDL1 induced >40% complete regressions and was superior to AAV-aPDL1, as well as to aPDL1 mAb given systemically or locally. SFV-aPDL1 induced abscopal effects and was also efficacious against B16-ovalbumin (OVA). The higher SFV-aPDL1 antitumor activity could be related to local upregulation of interferon-stimulated genes because of SFV RNA replication. This was confirmed by combining local SFV-LacZ administration and systemic aPDL1 mAb, which provided higher antitumor effects than each separated agent. SFV-aPDL1 promoted tumor-specific CD8 T cells infiltration in both tumor models. In MC38, SFV-aPDL1 upregulated co-stimulatory markers (CD137/OX40) in tumor CD8 T cells, and its combination with anti-CD137 mAb showed more pronounced antitumor effects than each single agent. These results indicate that local transient expression of immunomodulatory mAbs using non-propagative RNA vectors inducing type I interferon (IFN-I) responses represents a potent and safe approach for cancer treatment.


Subject(s)
Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Gene Expression , Genetic Vectors/genetics , Neoplasms/genetics , Neoplasms/immunology , RNA Viruses/genetics , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Line , Dependovirus/genetics , Disease Models, Animal , Female , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Humans , Immunomodulation/drug effects , Immunophenotyping , Injections, Intralesional , Mice , Neoplasms/pathology , Neoplasms/therapy , Recombinant Fusion Proteins/genetics , Semliki forest virus/genetics , Survival Rate , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Burden
7.
FASEB J ; 33(6): 7578-7587, 2019 06.
Article in English | MEDLINE | ID: mdl-30892966

ABSTRACT

Macrophages play a central role in tissue remodeling, repair, and resolution of inflammation. Macrophage polarization to M1 or M2 activation status may determine the progression or resolution of the inflammatory response. We have previously reported that cardiotrophin-1 (CT-1) displays both cytoprotective and metabolic activities. The role of CT-1 in inflammation remains poorly understood. Here, we employed recombinant CT-1 (rCT-1) and used CT-1-null mice and myeloid-specific CT-1 transgenic mice to investigate whether CT-1 might play a role in the modulation of the inflammatory response. We observed that CT-1 deficiency was associated with enhanced release of inflammatory mediators and with stronger activation of NF-κB in response to LPS, whereas the inflammatory response was attenuated in CT-1 transgenic mice or by administering rCT-1 to wild-type animals prior to LPS challenge. We found that CT-1 promoted IL-6 expression only by nonhematopoietic cells, whereas LPS up-regulated IL-6 expression in both hematopoietic and nonhematopoietic cells. Notably, rCT-1 inhibited LPS-mediated soluble IL-6R induction. Using IL-6-/- mice, we showed that rCT-1 inhibited LPS-induced TNF-α and IFN-γ response in an IL-6-independent manner. Importantly, we demonstrated that CT-1 primes macrophages for IL-4-dependent M2 polarization by inducing IL-4 receptor expression. Mechanistic analyses showed that the signal transducer and activator of transcription 3-suppressor of cytokine signaling 3 axis mediates this effect. Our findings support the notion that CT-1 is a critical regulator of inflammation and suggest that rCT-1 could be a molecule with potential therapeutic application in inflammatory conditions.-Carneros, D., Santamaría, E. M., Larequi, E., Vélez-Ortiz, J. M., Reboredo, M., Mancheño, U., Perugorria, M. J., Navas, P., Romero-Gómez, M., Prieto, J., Hervás-Stubbs, S., Bustos, M. Cardiotrophin-1 is an anti-inflammatory cytokine and promotes IL-4-induced M2 macrophage polarization.


Subject(s)
Cell Polarity , Cytokines/physiology , Inflammation/prevention & control , Interleukin-4/physiology , Macrophages/cytology , Animals , Interleukin-6/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic
8.
Cancer Res ; 77(13): 3672-3684, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28522749

ABSTRACT

Recent studies have found that tumor-infiltrating lymphocytes (TIL) expressing PD-1 can recognize autologous tumor cells, suggesting that cells derived from PD-1+ TILs can be used in adoptive T-cell therapy (ACT). However, no study thus far has evaluated the antitumor activity of PD-1-selected TILs in vivo In two mouse models of solid tumors, we show that PD-1 allows identification and isolation of tumor-specific TILs without previous knowledge of their antigen specificities. Importantly, despite the high proportion of tumor-reactive T cells present in bulk CD8 TILs before expansion, only T-cell products derived from sorted PD-1+, but not from PD-1- or bulk CD8 TILs, specifically recognized tumor cells. The fold expansion of PD-1+ CD8 TILs was 10 times lower than that of PD-1- cells, suggesting that outgrowth of PD-1- cells was the limiting factor in the tumor specificity of cells derived from bulk CD8 TILs. The highly differentiated state of PD-1+ cells was likely the main cause hampering ex vivo expansion of this subset. Moreover, PD-1 precisely identified marrow-infiltrating, myeloma-specific T cells in a mouse model of multiple myeloma. In vivo, only cells expanded from PD-1+ CD8 TILs contained tumor progression, and their efficacy was enhanced by PDL-1 blockade. Overall, our data provide a rationale for the use of PD-1-selected TILs in ACT. Cancer Res; 77(13); 3672-84. ©2017 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/biosynthesis , Animals , Cell Line, Tumor , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology
9.
Oncotarget ; 7(4): 4522-30, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26683225

ABSTRACT

TIM3 belongs to a family of receptors that are involved in T-cell exhaustion and Treg functions. The development of new therapeutic agents to block this type of receptors is opening a new avenue in cancer immunotherapy. There are currently several clinical trials ongoing to combine different immune-checkpoint blockades to improve the outcome of cancer patients. Among these combinations we should underline PD1:PDL1 axis and TIM3 blockade, which have shown very promising results in preclinical settings. Most of these types of therapeutic agents are protein cell-derived products, which, although broadly used in clinical settings, are still subject to important limitations. In this work we identify by HT-SELEX TIM3 non-antigenic oligonucleotide aptamers (TIM3Apt) that bind with high affinity and specificity to the extracellular motives of TIM3 on the cell surface. The TIM3Apt1 in its monomeric form displays a potent antagonist capacity on TIM3-expressing lymphocytes, determining the increase of IFN-γ secretion. In colon carcinoma tumor-bearing mice, the combinatorial treatment of TIM3Apt1 and PDL1-antibody blockade is synergistic with a remarkable antitumor effect. Immunotherapeutic aptamers could represent an attractive alternative to monoclonal antibodies, as they exhibit important advantages; namely, lower antigenicity, being chemically synthesized agents with a lower price of manufacture, providing higher malleability, and antidote availability.


Subject(s)
Aptamers, Nucleotide/pharmacology , Immunotherapy , Melanoma/therapy , Oligonucleotides, Antisense/pharmacology , SELEX Aptamer Technique/methods , Tissue Inhibitor of Metalloproteinase-3/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Flow Cytometry , Humans , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Protein Kinases , T-Lymphocytes, Regulatory/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Cancer Res ; 75(3): 497-507, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25527611

ABSTRACT

Host responses are increasingly considered important for the efficacious response to experimental cancer therapies that employ viral vectors, but little is known about the specific nature of host responses required. In this study, we investigated the role of host type I interferons (IFN-I) in the efficacy of virally delivered therapeutic genes. Specifically, we used a Semliki Forest virus encoding IL12 (SFV-IL12) based on its promise as an RNA viral vector for cancer treatment. Intratumoral injection of SFV-IL12 induced production of IFN-I as detected in serum. IFN-I production was abolished in mice deficient for the IFNß transcriptional regulator IPS-1 and partially attenuated in mice deficient for the IFNß signaling protein TRIF. Use of bone marrow chimeric hosts established that both hematopoietic and stromal cells were involved in IFN-I production. Macrophages, plasmacytoid, and conventional dendritic cells were each implicated based on cell depletion experiments. Further, mice deficient in the IFN-I receptor (IFNAR) abolished the therapeutic activity of SFV-IL12, as did a specific antibody-mediated blockade of IFNAR signaling. Reduced efficacy was not caused by an impairment in IL12 expression, because IFNAR-deficient mice expressed the viral IL12 transgene even more strongly than wild-type (WT) hosts. Chimeric host analysis for the IFNAR involvement established a strict requirement in hematopoietic cells. Notably, although tumor-specific CD8 T lymphocytes expanded robustly after intratumoral injection of WT mice with SFV-IL12, this did not occur in mice where IFNAR was inactivated genetically or pharmacologically. Overall, our results argued that the antitumor efficacy of a virally based transgene therapeutic relied strongly on a vector-induced IFN-I response, revealing an unexpected mechanism of action that is relevant to a broad array of current translational products in cancer research.


Subject(s)
Antineoplastic Agents/chemistry , Genetic Therapy/methods , Genetic Vectors , Interleukin-12/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Crosses, Genetic , Dendritic Cells/cytology , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Immunotherapy , Interferon-alpha/metabolism , Interferon-beta/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , RNA, Viral/genetics , Recombinant Proteins/metabolism , Transgenes
11.
Liver Int ; 35(4): 1274-89, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24754307

ABSTRACT

BACKGROUND & AIMS: Adenoviral (Ad) vectors are currently one of the most efficient tools for in vivo gene transfer to the liver. However, anti-Ad immune responses limit the safety and efficacy of these vectors. The initial inflammatory reaction is a concern in terms of toxicity, and it favours the development of cellular and humoral responses leading to short transgene persistence and inefficient vector re-administrations. Therefore, safe and simple ways to interfere with these processes are needed. Study ways to deplete specific immune cell populations and their impact on liver-directed gene transfer. METHODS: First-generation Ad vectors encoding reporter genes (luciferase or ß-galactosidase) were injected intravenously into Balb/c mice. Kupffer cells and splenic macrophages were depleted by intravenous administration of clodronate liposomes. B lymphocytes, CD4(+) , CD8(+) T lymphocytes or NK cells were depleted by intraperitoneal injection of anti-M plus anti-D, anti-CD4, anti-CD8 or anti-asialo-GM1 antibodies respectively. Long-term evolution of luciferase expression in the liver was monitored by bioluminescence imaging. RESULTS: The anti-CD4 monoclonal antibody impaired cellular and humoral immune responses, leading to efficient vector re-administration. Clodronate liposomes had no impact on humoral responses but caused a 100-1000 fold increase in liver transduction, stabilized transgene expression, reduced the concentration of inflammatory cytokines, and inhibited lymphocyte activation. CONCLUSIONS: Transient CD4(+) T-cell depletion using antibodies is a clinically feasible procedure that allows efficient Ad redosing. Systemic administration of clodronate liposomes may further increase the safety and efficacy of vectors.


Subject(s)
Adenoviridae/immunology , CD4-Positive T-Lymphocytes/drug effects , Genetic Vectors , Immunosuppressive Agents/pharmacology , Liver/drug effects , Lymphocyte Depletion/methods , Transduction, Genetic , Transgenes , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Antibodies/pharmacology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Clodronic Acid/pharmacology , Female , Gene Expression Regulation , Genes, Reporter , Immunity, Humoral/drug effects , Liver/immunology , Liver/metabolism , Luciferases/biosynthesis , Luciferases/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Time Factors , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
12.
J Immunol ; 193(3): 1151-61, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24973449

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are considered to be the principal type-I IFN (IFN-I) source in response to viruses, whereas the contribution of conventional DCs (cDCs) has been underestimated because, on a per-cell basis, they are not considered professional IFN-I-producing cells. We have investigated their respective roles in the IFN-I response required for CTL activation. Using a nonreplicative virus, baculovirus, we show that despite the high IFN-I-producing abilities of pDCs, in vivo cDCs but not pDCs are the pivotal IFN-I producers upon viral injection, as demonstrated by selective pDC or cDC depletion. The pathway involved in the virus-triggered IFN-I response is dependent on TLR9/MyD88 in pDCs and on stimulator of IFN genes (STING) in cDCs. Importantly, STING is the key molecule for the systemic baculovirus-induced IFN-I response required for CTL priming. The supremacy of cDCs over pDCs in fostering the IFN-I response required for CTL activation was also verified in the lymphocytic choriomeningitis virus model, in which IFN-ß promoter stimulator 1 plays the role of STING. However, when the TLR-independent virus-triggered IFN-I production is impaired, the pDC-induced IFNs-I have a primary impact on CTL activation, as shown by the detrimental effect of pDC depletion and IFN-I signaling blockade on the residual lymphocytic choriomeningitis virus-triggered CTL response detected in IFN-ß promoter stimulator 1(-/-) mice. Our findings reveal that cDCs play a major role in the TLR-independent virus-triggered IFN-I production required for CTL priming, whereas pDC-induced IFNs-I are dispensable but become relevant when the TLR-independent IFN-I response is impaired.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Dendritic Cells/virology , Interferon Type I/biosynthesis , Nucleopolyhedroviruses/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/classification , Female , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/physiology , Signal Transduction/immunology , Toll-Like Receptor 9/physiology
13.
J Immunol ; 189(7): 3299-310, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22925929

ABSTRACT

Previous mouse and human studies have demonstrated that direct IFN-α/ß signaling on naive CD8 T cells is critical to support their expansion and acquisition of effector functions. In this study, we show that human naive CD8 T cells primed in the presence of IFN-α possess a heightened ability to respond to homeostatic cytokines and to secondary Ag stimulation, but rather than differentiating to effector or memory CTLs, they preserve nature-like phenotypic features. These are qualities associated with greater efficacy in adoptive immunotherapy. In a mouse model of adoptive transfer, CD8 T cells primed in the presence of IFN-α are able to persist and to mediate a robust recall response even after a long period of naturally driven homeostatic maintenance. The long-lasting persistence of IFN-α-primed CD8 T cells is favored by their enhanced responsiveness to IL-15 and IL-7, as demonstrated in IL-15(-/-) and IL-7(-/-) recipient mice. In humans, exposure to IFN-α during in vitro priming of naive HLA-A2(+) CD8 T cells with autologous dendritic cells loaded with MART1(26-35) peptide renders CD8 T cells with an improved capacity to respond to homeostatic cytokines and to specifically lyse MART1-expressing melanoma cells. Furthermore, in a mouse model of melanoma, adoptive transfer of tumor-specific CD8 T cells primed ex vivo in the presence of IFN-α exhibits an improved ability to contain tumor progression. Therefore, exposure to IFN-α during priming of naive CD8 T cells imprints decisive information on the expanded cells that can be exploited to improve the efficacy of adoptive T cell therapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytokines/physiology , Homeostasis/immunology , Immunization, Secondary/methods , Immunologic Memory , Interferon-alpha/physiology , Lymphocyte Activation/immunology , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer/methods , Animals , Antigens/physiology , CD8-Positive T-Lymphocytes/transplantation , Cells, Cultured , Humans , Interleukin-15/physiology , Interleukin-17/physiology , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/transplantation
14.
J Transl Med ; 10: 122, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22704060

ABSTRACT

BACKGROUND: Adeno-associated vectors (rAAV) have been used to attain long-term liver gene expression. In humans, the cellular immune response poses a serious obstacle for transgene persistence while neutralizing humoral immunity curtails re-administration. Porphobilinogen deaminase (PBGD) haploinsufficiency (acute intermittent porphyria) benefits from liver gene transfer in mouse models and clinical trials are about to begin. In this work, we sought to study in non-human primates the feasibility of repeated gene-transfer with intravenous administration of rAAV5 vectors under the effects of an intensive immunosuppressive regimen and to analyze its ability to circumvent T-cell immunity and thereby prolong transgene expression. METHODS: Three female Macaca fascicularis were intravenously injected with 1 x 10(13) genome copies/kg of rAAV5 encoding the human PBGD. Mycophenolate mofetil (MMF), anti-thymocyte immunoglobulin, methylprednisolone, tacrolimus and rituximab were given in combination during 12 weeks to block T- and B-cell mediated adaptive immune responses in two macaques. Immunodeficient and immunocompetent mice were intravenously injected with 5 x 10(12) genome copies/kg of rAAV5-encoding luciferase protein. Forty days later MMF, tacrolimus and rituximab were daily administrated to ascertain whether the immunosuppressants or their metabolites could interfere with transgene expression. RESULTS: Macaques given a rAAV5 vector encoding human PBGD developed cellular and humoral immunity against viral capsids but not towards the transgene. Anti-AAV humoral responses were attenuated during 12 weeks but intensely rebounded following cessation of the immunosuppressants. Accordingly, subsequent gene transfer with a rAAV5 vector encoding green fluorescent protein was impossible. One macaque showed enhanced PBGD expression 25 weeks after rAAV5-pbgd administration but overexpression had not been detected while the animal was under immunosuppression. As a potential explanation, MMF decreases transgene expression in mouse livers that had been successfully transduced by a rAAV5 several weeks before MMF onset. Such a silencing effect was independent of AAV complementary strand synthesis and requires an adaptive immune system. CONCLUSIONS: These results indicate that our transient and intensive pharmacological immunosuppression fails to improve AAV5-based liver gene transfer in non-human primates. The reasons include an incomplete restraint of humoral immune responses to viral capsids that interfere with repeated gene transfer in addition to an intriguing MMF-dependent drug-mediated interference with liver transgene expression.


Subject(s)
Dependovirus/metabolism , Gene Transfer Techniques , Immunosuppressive Agents/pharmacology , Liver/metabolism , Macaca fascicularis/immunology , Animals , Antigens, Viral/immunology , Capsid/immunology , DNA, Viral/blood , Dependovirus/drug effects , Female , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Immunity/drug effects , Immunity, Humoral/drug effects , Immunity, Humoral/immunology , Immunosuppression Therapy , Immunosuppressive Agents/administration & dosage , Injections, Intravenous , Liver/drug effects , Mice , Mice, Transgenic , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/pharmacology , Serotyping , Transgenes/genetics , Treatment Failure
15.
Gut ; 60(3): 341-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20855451

ABSTRACT

BACKGROUND AND AIMS: New options are needed for the management and prevention of colorectal cancer liver metastases. Interleukin 12 (IL-12) is an immunostimulatory cytokine with proven antitumour effect in animal models. Despite evidence indicating its biological effect in humans, neither the recombinant protein nor gene therapy vectors expressing IL-12 have shown a relevant benefit in patients with cancer. OBJECTIVE: To develop a new approach to overcome the difficulties in obtaining a suitable expression pattern and the immunosuppressive milieu in the tumours which contribute to this poor performance. METHODS: A high-capacity ('gutless') adenoviral vector carrying a liver-specific, mifepristone (Mif)-inducible system for the expression of IL-12 (HC-Ad/RUmIL-12) was used in combination with chemotherapy. Tumours were established in the liver of C57BL/6 mice by inoculation of MC38 colon cancer cells. RESULTS: Intrahepatic injection of HC-Ad/RUmIL-12 and tailored induction regimens allowed the maintenance of safe and efficient levels of IL-12 in vivo. An individualised, stepwise increase in the dose of Mif (125-4000 µg/kg) was needed to compensate for the progressive but transient downregulation of the inducible system. Repeated cycles of Mif induction (every 24 h for 10 days) were needed for optimal tumour eradication. However, complete protection against tumour rechallenge was seen in < 25% of the animals. The administration of oxaliplatin (5 mg/kg intraperitoneally) 3 days before starting the induction regimen achieved efficient elimination of liver metastases with a single cycle of IL-12 induction, and improved protection against tumour rechallenge. This was associated with a shift in the tumour microenvironment towards a more pro-immunogenic phenotype, with an increase in the CD8+/T regulatory cell ratio and a reduction in myeloid-derived suppressor cells. These effects were not seen with 5-fluorouracil, irinotecan or gemcitabine. CONCLUSIONS: Long-term controlled expression of IL-12 using an HC-Ad vector in combination with oxaliplatin is effective and clinically applicable against hepatic colon cancer metastases.


Subject(s)
Antineoplastic Agents/therapeutic use , Genetic Therapy/methods , Interleukin-12/biosynthesis , Liver Neoplasms/secondary , Organoplatinum Compounds/therapeutic use , Animals , Colorectal Neoplasms/immunology , Combined Modality Therapy , Down-Regulation/drug effects , Female , Genetic Vectors , Immune Tolerance/drug effects , Interleukin-12/genetics , Liver/metabolism , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Mice , Mice, Inbred C57BL , Mice, Nude , Mifepristone/pharmacology , Oxaliplatin , Transduction, Genetic , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
16.
Eur J Immunol ; 40(12): 3389-402, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21108462

ABSTRACT

IFN-α/ß link innate and adaptive immune responses by directly acting on naïve CD8(+) T cells. This concept unveiled in mice remains unexplored in humans. To investigate that, human CD8(+) CD45RO(-) cells were stimulated with beads coated with anti-CD3 and anti-CD28 mAb, mimicking Ag (type-1) and co-stimulatory (type-2) signals, in the presence or absence of IFN-α and their transcriptional profiles were defined by cDNA-microarrays. We show that IFN-α provides a strong third signal directly to human CD8(+) T cells resulting in regulation of critical genes for their overall activation. This transcriptional effect was substantiated at the protein level and verified by functional assays. Interestingly, the biological effects derived from this stimulation vary depending on the CD8(+) T-cell population. Thus, whereas IFN-α increases the proliferative capacity of naïve CD8(+) T cells, it inhibits or does not affect the proliferation of Ag-experienced cells, such as memory and effector CTL, including CMV-specific lymphocytes. Cytolysis and IFN-γ-secretion of all these populations are enhanced by IFN-α-derived signals, which are critical in naïve CD8(+) T cells for acquisition of effector functions. Our findings in human CD8(+) T cells are informative to understand and improve IFN-α-based therapies for viral and malignant diseases.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Interferon-alpha/pharmacology , Interferon-gamma/biosynthesis , Phosphoproteins/immunology , T-Lymphocyte Subsets/drug effects , Viral Matrix Proteins/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Gene Expression Profiling , Humans , Immunization , Immunologic Memory/drug effects , Interferon-gamma/genetics , Oligonucleotide Array Sequence Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
17.
Mol Ther ; 18(4): 754-65, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20087317

ABSTRACT

Repeated administration of gene therapies is hampered by host immunity toward vectors and transgenes. Attempts to circumvent antivector immunity include pharmacological immunosuppression or alternating different vectors and vector serotypes with the same transgene. Our studies show that B-cell depletion with anti-CD20 monoclonal antibody and concomitant T-cell inhibition with clinically available drugs permits repeated liver gene transfer to a limited number of nonhuman primates with recombinant adenovirus. Adenoviral vector-mediated transfer of the herpes simplex virus type 1 thymidine kinase (HSV1-tk) reporter gene was visualized in vivo with a semiquantitative transgene-specific positron emission tomography (PET) technique, liver immunohistochemistry, and immunoblot for the reporter transgene in needle biopsies. Neutralizing antibody and T cell-mediated responses toward the viral capsids were sequentially monitored and found to be repressed by the drug combinations tested. Repeated liver transfer of the HSV1-tk reporter gene with the same recombinant adenoviral vector was achieved in macaques undergoing a clinically feasible immunosuppressive treatment that ablated humoral and cellular immune responses. This strategy allows measurable gene retransfer to the liver as late as 15 months following the first adenoviral exposure in a macaque, which has undergone a total of four treatments with the same adenoviral vector.


Subject(s)
Adenoviridae/immunology , Antibodies, Monoclonal/administration & dosage , Gene Transfer Techniques , Genetic Vectors/immunology , Immunosuppressive Agents/administration & dosage , Liver/immunology , Lymphocyte Depletion , Adenoviridae/genetics , Animals , Antibodies, Monoclonal, Murine-Derived , Antibodies, Neutralizing/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Female , Genetic Therapy , Genetic Vectors/genetics , Macaca , Reassortant Viruses , Recombination, Genetic , Rituximab , T-Lymphocytes/immunology , Tacrolimus/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...