Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cancer Immunol Res ; 8(1): 70-80, 2020 01.
Article in English | MEDLINE | ID: mdl-31699709

ABSTRACT

Given its ability to induce both humoral and cellular immune responses, NY-ESO-1 has been considered a suitable antigen for a cancer vaccine. Despite promising results from early-phase clinical studies in patients with melanoma, NY-ESO-1 vaccine immunotherapy has not been widely investigated in larger trials; consequently, many questions remain as to the optimal vaccine formulation, predictive biomarkers, and sequencing and timing of vaccines in melanoma treatment. We conducted an adjuvant phase I/II clinical trial in high-risk resected melanoma to optimize the delivery of poly-ICLC, a TLR-3/MDA-5 agonist, as a component of vaccine formulation. A phase I dose-escalation part was undertaken to identify the MTD of poly-ICLC administered in combination with NY-ESO-1 and montanide. This was followed by a randomized phase II part investigating the MTD of poly-ICLC with NY-ESO-1 with or without montanide. The vaccine regimens were generally well tolerated, with no treatment-related grade 3/4 adverse events. Both regimens induced integrated NY-ESO-1-specific CD4+ T-cell and humoral responses. CD8+ T-cell responses were mainly detected in patients receiving montanide. T-cell avidity toward NY-ESO-1 peptides was higher in patients vaccinated with montanide. In conclusion, NY-ESO-1 protein in combination with poly-ICLC is safe, well tolerated, and capable of inducing integrated antibody and CD4+ T-cell responses in most patients. Combination with montanide enhances antigen-specific T-cell avidity and CD8+ T-cell cross-priming in a fraction of patients, indicating that montanide contributes to the induction of specific CD8+ T-cell responses to NY-ESO-1.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/therapeutic use , Carboxymethylcellulose Sodium/analogs & derivatives , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Mannitol/analogs & derivatives , Melanoma/immunology , Membrane Proteins/administration & dosage , Oleic Acids/administration & dosage , Poly I-C/administration & dosage , Polylysine/analogs & derivatives , Adjuvants, Immunologic/administration & dosage , Aged , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Carboxymethylcellulose Sodium/administration & dosage , Cross-Priming/immunology , Female , Humans , Interferon Inducers/administration & dosage , Male , Mannitol/administration & dosage , Melanoma/therapy , Membrane Proteins/immunology , Middle Aged , Patient Safety , Polylysine/administration & dosage , Skin Neoplasms/immunology , Skin Neoplasms/therapy , Treatment Outcome
2.
Front Immunol ; 10: 725, 2019.
Article in English | MEDLINE | ID: mdl-31024557

ABSTRACT

Objective: Toll-like receptor-3 agonist Poly-ICLC has been known to activate immune cells and induce HIV replication in pre-clinical experiments. In this study we investigated if Poly-ICLC could be used for disrupting HIV latency while simultaneously enhancing innate immune responses. Design: This was a randomized, placebo-controlled, double-blinded trial in aviremic, cART-treated HIV-infected subjects. Participants (n = 15) were randomized 3:1 to receive two consecutive daily doses of Poly-ICLC (1.4 mg subcutaneously) vs. placebo. Subjects were observed for adverse events, immune activation, and viral replication. Methods: Besides primary outcomes of safety and tolerability, several longitudinal immune parameters were evaluated including immune cell phenotype and function via flowcytometry, ELISA, and transcriptional profiling. PCR assays for plasma HIV-1 RNA, CD4+ T cell-associated HIV-1 RNA, and proviral DNA were performed to measure HIV reservoirs and latency. Results: Poly-ICLC was overall safe and well-tolerated. Poly-ICLC-related adverse events were Grade 1/2, with the exception of one Grade 3 neutropenia which was short-lived. Mild Injection site reactions were observed in nearly all participants in the Poly-ICLC arm. Transcriptional analyses revealed upregulation of innate immune pathways in PBMCs following Poly-ICLC treatment, including strong interferon signaling accompanied by transient increases in circulating IP-10 (CXCL10) levels. These responses generally peaked by 24-48 h after the first injection and returned to baseline by day 8. CD4+ T cell number and phenotype were unchanged, plasma viral control was maintained and no significant effect on HIV reservoirs was observed. Conclusions: These finding suggest that Poly-ICLC could be safely used for inducing transient innate immune responses in treated HIV+ subjects indicating promise as an adjuvant for HIV therapeutic vaccines. Trial Registration: www.ClinicalTrials.gov, identifier: NCT02071095.


Subject(s)
Carboxymethylcellulose Sodium/analogs & derivatives , HIV Infections/drug therapy , HIV Infections/immunology , Immunity, Innate/drug effects , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Toll-Like Receptor 3/metabolism , Adult , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Carboxymethylcellulose Sodium/therapeutic use , Double-Blind Method , Female , HIV/immunology , HIV Infections/metabolism , Humans , Male , Middle Aged , Polylysine/therapeutic use
3.
Clin Cancer Res ; 24(20): 4937-4948, 2018 10 15.
Article in English | MEDLINE | ID: mdl-29950349

ABSTRACT

Purpose: Polyinosinic-polycytidylic acid-poly-l-lysine carboxymethylcellulose (poly-ICLC), a synthetic double-stranded RNA complex, is a ligand for toll-like receptor-3 and MDA-5 that can activate immune cells, such as dendritic cells, and trigger natural killer cells to kill tumor cells.Patients and Methods: In this pilot study, eligible patients included those with recurrent metastatic disease in whom prior systemic therapy (head and neck squamous cell cancer and melanoma) failed. Patients received 2 treatment cycles, each cycle consisting of 1 mg poly-ICLC 3× weekly intratumorally (IT) for 2 weeks followed by intramuscular (IM) boosters biweekly for 7 weeks, with a 1-week rest period. Immune response was evaluated by immunohistochemistry (IHC) and RNA sequencing (RNA-seq) in tumor and blood.Results: Two patients completed 2 cycles of IT treatments, and 1 achieved clinical benefit (stable disease, progression-free survival 6 months), whereas the remainder had progressive disease. Poly-ICLC was well tolerated, with principal side effects of fatigue and inflammation at injection site (

Subject(s)
Carboxymethylcellulose Sodium/analogs & derivatives , Immunologic Factors/administration & dosage , Immunomodulation/drug effects , Neoplasms/drug therapy , Neoplasms/immunology , Poly I-C/administration & dosage , Polylysine/analogs & derivatives , Aged , Biopsy , Carboxymethylcellulose Sodium/administration & dosage , Carboxymethylcellulose Sodium/adverse effects , Drug Administration Schedule , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Immunologic Factors/adverse effects , Injections, Intralesional , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasms/diagnosis , Neoplasms/mortality , Pilot Projects , Poly I-C/adverse effects , Polylysine/administration & dosage , Polylysine/adverse effects , Prognosis , Tomography, X-Ray Computed , Treatment Outcome
4.
J Immunother Cancer ; 6(1): 38, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29773080

ABSTRACT

BACKGROUND: Cancer-testis antigen NY-ESO-1 is a highly immunogenic melanoma antigen which has been incorporated into adjuvant vaccine clinical trials. Three such early-phase trials were conducted at our center among patients with high-risk resected melanoma. We herein report on the pooled long-term survival outcomes of these patients in comparison to historical controls. METHODS: All melanoma patients treated at NYU Langone Health under any of three prospective adjuvant NY-ESO-1 vaccine trials were retrospectively pooled into a single cohort. All such patients with stage III melanoma were subsequently compared to historical control patients identified via a prospective institutional database with protocol-driven follow-up. Survival times were calculated using the Kaplan-Meier method, and Cox proportional hazard models were employed to identify significant prognostic factors and control for confounding variables. RESULTS: A total of 91 patients were treated with an NY-ESO-1 vaccine for the treatment of high-risk resected melanoma. Of this group, 67 patients were stage III and were selected for comparative analysis with 123 historical control patients with resected stage III melanoma who received no adjuvant therapy. Among the pooled vaccine cohort (median follow-up 61 months), the estimated median recurrence-free survival was 45 months, while the median overall survival was not yet reached. In the control cohort of 123 patients (median follow-up 30 months), the estimated median recurrence-free and overall survival were 22 and 58 months, respectively. Within the retrospective stage III cohort, NY-ESO-1 vaccine was associated with decreased risk of recurrence (HR = 0.56, p < 0.01) and death (HR = 0.51, p = 0.01). Upon controlling for sub-stage, the adjuvant NY-ESO-1 clinical trial cohort continued to exhibit decreased risk of recurrence (HR = 0.45, p < 0.01) and death (HR = 0.40, p < 0.01). CONCLUSIONS: In this small retrospective cohort of resected stage III melanoma patients, adjuvant NY-ESO-1 vaccine immunotherapy was associated with longer recurrence-free and overall survival relative to historical controls. These data support the continued investigation of adjuvant NY-ESO-1 based immunotherapy regimens in melanoma.


Subject(s)
Cancer Vaccines/therapeutic use , Chemotherapy, Adjuvant/methods , Immunotherapy/methods , Melanoma/drug therapy , Cancer Vaccines/pharmacology , Female , Humans , Male , Melanoma/pathology , Middle Aged
5.
Cell Res ; 27(1): 74-95, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28025976

ABSTRACT

Immunotherapy using dendritic cell (DC)-based vaccination is an approved approach for harnessing the potential of a patient's own immune system to eliminate tumor cells in metastatic hormone-refractory cancer. Overall, although many DC vaccines have been tested in the clinic and proven to be immunogenic, and in some cases associated with clinical outcome, there remains no consensus on how to manufacture DC vaccines. In this review we will discuss what has been learned thus far about human DC biology from clinical studies, and how current approaches to apply DC vaccines in the clinic could be improved to enhance anti-tumor immunity.


Subject(s)
Dendritic Cells/immunology , Immunotherapy , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Dendritic Cells/cytology , Humans , Immunity , Vaccination
6.
Methods Mol Biol ; 1403: 763-77, 2016.
Article in English | MEDLINE | ID: mdl-27076166

ABSTRACT

Exploitation of the patient's own immune system to induce antitumor immune responses using dendritic cell (DC) immunotherapy has been established in early clinical trials as a safe and promising therapeutic approach for cancer. However, their limited success in larger clinical trials highlights the need to optimize DC vaccine preparations. This chapter describes the methodologies utilized for the preparation of the DC vaccine most commonly used in clinical trials. Optional variations at different stages in DC vaccine preparation, based on the nature of antigen, delivery of antigen, maturation stimuli, and mode of administration for DC vaccines, are also presented for consideration as these are often dependent on the disease setting, desired immune response, and/or resources available.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Cell Culture Techniques , Cell Separation , Clinical Trials as Topic , Cytokines/pharmacology , Dendritic Cells/drug effects , Humans , Immunotherapy
8.
Cancer Immunol Res ; 3(3): 278-287, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25633712

ABSTRACT

The Toll-like receptor (TLR) 7/8 agonist resiquimod has been used as an immune adjuvant in cancer vaccines. We evaluated the safety and immunogenicity of the cancer testis antigen NY-ESO-1 given in combination with Montanide (Seppic) with or without resiquimod in patients with high-risk melanoma. In part I of the study, patients received 100 µg of full-length NY-ESO-1 protein emulsified in 1.25 mL of Montanide (day 1) followed by topical application of 1,000 mg of 0.2% resiquimod gel on days 1 and 3 (cohort 1) versus days 1, 3, and 5 (cohort 2) of a 21-day cycle. In part II, patients were randomized to receive 100-µg NY-ESO-1 protein plus Montanide (day 1) followed by topical application of placebo gel [(arm A; n = 8) or 1,000 mg of 0.2% resiquimod gel (arm B; n = 12)] using the dosing regimen established in part I. The vaccine regimens were generally well tolerated. NY-ESO-1-specific humoral responses were induced or boosted in all patients, many of whom had high titer antibodies. In part II, 16 of 20 patients in both arms had NY-ESO-1-specific CD4⁺ T-cell responses. CD8⁺ T-cell responses were only seen in 3 of 12 patients in arm B. Patients with TLR7 SNP rs179008 had a greater likelihood of developing NY-ESO-1-specific CD8⁺ responses. In conclusion, NY-ESO-1 protein in combination with Montanide with or without topical resiquimod is safe and induces both antibody and CD4⁺ T-cell responses in the majority of patients; the small proportion of CD8⁺ T-cell responses suggests that the addition of topical resiquimod to Montanide is not sufficient to induce consistent NY-ESO-1-specific CD8⁺ T-cell responses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Imidazoles/immunology , Melanoma/therapy , Membrane Proteins/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Neoplasm/therapeutic use , Antibody Formation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Immunity, Cellular , Male , Mannitol/administration & dosage , Mannitol/analogs & derivatives , Middle Aged , Oleic Acids/administration & dosage , Peptide Fragments/immunology , Vaccination
9.
Vaccine ; 33(2): 388-95, 2015 Jan 03.
Article in English | MEDLINE | ID: mdl-25444812

ABSTRACT

Therapeutic interventions for HIV-1 that successfully augment adaptive immunity to promote killing of infected cells may be a requisite component of strategies to reduce latent cellular reservoirs. Adoptive immunotherapies utilizing autologous monocyte-derived dendritic cells (DCs) that have been activated and antigen loaded ex vivo may serve to circumvent defects in DC function that are present during HIV infection in order to enhance adaptive immune responses. Here we detail the clinical preparation of DCs loaded with autologous aldrithiol-2 (AT-2)-inactivated HIV that have been potently activated with the viral mimic, Polyinosinic-polycytidylic acid-poly-l-lysine carboxymethylcellulose (Poly-ICLC). HIV is first propagated from CD4+ T cells from HIV-infected donors and then rendered non-replicative by chemical inactivation with aldrithiol-2 (AT-2), purified, and quantified. Viral inactivation is confirmed through measurement of Tat-regulated ß-galactosidase reporter gene expression following infection of TZM-bl cells. In-process testing for sterility, mycoplasma, LPS, adventitious agents, and removal of AT-2 is performed on viral preparations. Autologous DCs are generated and pulsed with autologous AT-2-inactivated virus and simultaneously stimulated with Poly-ICLC to constitute the final DC vaccine product. Phenotypic identity, maturation, and induction of HIV-specific adaptive immune responses are confirmed via flow cytometric analysis of DCs and cocultured autologous CD4+ and CD8+ T cells. Lot release criteria for the DC vaccine have been defined in accordance with Good Manufacturing Practice (GMP) guidelines. The demonstrated feasibility of this approach has resulted in approval by the FDA for investigational use in antiretroviral (ART) suppressed individuals. We discuss how this optimized DC formulation may enhance the quality of anti-HIV adaptive responses beyond what has been previously observed during DC immunotherapy trials for HIV infection.


Subject(s)
AIDS Vaccines/chemistry , AIDS Vaccines/immunology , Adaptive Immunity , Carboxymethylcellulose Sodium/analogs & derivatives , Dendritic Cells/immunology , HIV-1/immunology , Poly I-C/immunology , Polylysine/analogs & derivatives , 2,2'-Dipyridyl/analogs & derivatives , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Disulfides , HIV Infections/immunology , HIV Infections/therapy , HIV-1/growth & development , HIV-1/isolation & purification , HIV-1/physiology , Humans , Immunotherapy, Adoptive/methods , Polylysine/immunology , Virus Inactivation , beta-Galactosidase/genetics
10.
J Immunother Cancer ; 2(1): 32, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-28837000

ABSTRACT

Studies suggest that conventional cancer therapies given after immunotherapy (IT) can boost antitumor immunity and possibly improve response rates and progression-free survival. We report two cases of metastatic breast cancer with durable complete responses (CRs) after sequential IT and endocrine therapy. Immune analyses of these long-term disease-free breast cancer patients previously treated with imiquimod (IMQ) suggest in-situ vaccination is achieved by topical application of the TLR-7 agonist directly onto tumors. Furthermore, IT-induced antigen-specific T cells were expanded by subsequent endocrine therapy and correlated with response, persisting > 2 years. Our findings therefore suggest that the induction/boosting of polyfunctional tumor antigen-specific T in response to sequential immune endocrine therapy and detected directly ex-vivo can serve as a peripheral blood biomarker for true clinical benefit.

11.
J Vis Exp ; (78)2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23928481

ABSTRACT

While clinical studies have established that antigen-loaded DC vaccines are safe and promising therapy for tumors, their clinical efficacy remains to be established. The method described below, prepared in accordance with Good Manufacturing Process (GMP) guidelines, is an optimization of the most common ex vivo preparation method for generating large numbers of DCs for clinical studies. Our method utilizes the synthetic TLR 3 agonist Polyinosinic-Polycytidylic Acid-poly-L-lysine Carboxymethylcellulose (Poly-ICLC) to stimulate the DCs. Our previous study established that Poly-ICLC is the most potent individual maturation stimulus for human DCs as assessed by an upregulation of CD83 and CD86, induction of interleukin-12 (IL-12), tumor necrosis factor (TNF), interferon gamma-induced protein 10 (IP-10), interleukmin 1 (IL-1), and type I interferons (IFN), and minimal interleukin 10 (IL-10) production. DCs are differentiated from frozen peripheral blood mononuclear cells (PBMCs) obtained by leukapheresis. PBMCs are isolated by Ficoll gradient centrifugation and frozen in aliquots. On Day 1, PBMCs are thawed and plated onto tissue culture flasks to select for monocytes which adhere to the plastic surface after 1-2 hr incubation at 37 °C in the tissue culture incubator. After incubation, the lymphocytes are washed off and the adherent monocytes are cultured for 5 days in the presence of interleukin-4 (IL-4) and granulocyte macrophage-colony stimulating factor (GM-CSF) to differentiate to immature DCs. On Day 6, immature DCs are pulsed with the keyhole limpet hemocyanin (KLH) protein which serves as a control for the quality of the vaccine and may boost the immunogenicity of the vaccine. The DCs are stimulated to mature, loaded with peptide antigens, and incubated overnight. On Day 7, the cells are washed, and frozen in 1 ml aliquots containing 4-20 x 10(6) cells using a controlled-rate freezer. Lot release testing for the batches of DCs is performed and must meet minimum specifications before they are injected into patients.


Subject(s)
Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Antigens, Neoplasm , Carboxymethylcellulose Sodium/analogs & derivatives , Carboxymethylcellulose Sodium/pharmacology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-4/pharmacology , Leukocytes, Mononuclear/cytology , Poly I-C/pharmacology , Polylysine/analogs & derivatives , Polylysine/pharmacology
12.
Ann N Y Acad Sci ; 1284: 31-45, 2013 May.
Article in English | MEDLINE | ID: mdl-23651191

ABSTRACT

The U.S. Food and Drug Administration's approval of the first cell-based immunotherapy has rejuvenated interest in the field. Early clinical trials have established the ability of dendritic cell (DC) immunotherapy to exploit a patient's own immune system to induce antitumor immune responses. However, suboptimal conditions for generating potent immunostimulatory DCs, in addition to the suppression mediated by the tumor microenvironment, have contributed to limited clinical success in vivo. Therefore, combining DC vaccines with new approaches that enhance immunogenicity and overcome the regulatory mechanisms underlying peripheral tolerance may be key to achieving effective, durable, antitumor immune responses that translate to better clinical outcomes.


Subject(s)
Antigen-Presenting Cells/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Immunotherapy/methods , Animals , Antigens/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Humans
13.
J Acquir Immune Defic Syndr ; 61(5): 535-44, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22902724

ABSTRACT

OBJECTIVE: Myeloid dendritic cell (mDC) dysfunction during HIV infection may hinder the formation of both innate and adaptive immune responses and contribute to pathogenesis. Our objective was to determine whether circulating factors during chronic HIV infection impair mDC function with respect to secretion of IL-12, a pro-Th1 cytokine, and T-cell stimulatory capacity. Particular focus was placed on the effect of combination antiretroviral therapy (cART) and the role of HIV itself on mDC function. METHODS: Monocyte-derived DC (moDC) from uninfected donors were exposed to plasma from HIV-infected individuals before Toll-like receptor (TLR) stimulation. Cytokine secretion was measured via cytokine bead arrays, and T-cell proliferation and IFNγ secretion was evaluated after coculture with naive CD4 T cells. Expression of genes central to TLR-mediated signal transduction was analyzed via quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) arrays and western blot. RESULTS: Exposure of monocyte-derived DC to plasma from untreated HIV-infected donors suppressed secretion of IL-12, and impaired Th1-skewing of CD4 T cells. The suppressive effect was less by plasma donors receiving cART. Removal of virus from plasma did not relieve suppression nor was IL-12 secretion decreased on addition of HIV to control plasma. On a transcriptional level, decreased expression of IKKß, a key regulator in the TLR/NF-kappaB signaling pathway, corresponded to suppressed cytokine secretion. CONCLUSIONS: Plasma factors during chronic HIV infection impair mDC function in a manner that likely impacts the formation of immune responses to HIV, opportunistic pathogens, and vaccines. Despite partial alleviation by cART, this suppression was not directly mediated by HIV.


Subject(s)
Dendritic Cells/immunology , HIV Infections/blood , HIV Infections/immunology , HIV-1 , Interleukin-12/biosynthesis , Adult , CD4-Positive T-Lymphocytes/immunology , Coculture Techniques , Cross-Sectional Studies , Female , Gene Expression , HIV Infections/drug therapy , HIV Infections/genetics , HIV-1/immunology , Humans , I-kappa B Kinase/genetics , Immune Tolerance , Male , Middle Aged , Signal Transduction , Th1 Cells/immunology , Viral Load/immunology , Young Adult
14.
J Clin Invest ; 121(3): 1088-101, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21339641

ABSTRACT

Plasmacytoid DCs (pDCs) are innate immune cells that are specialized to produce IFN-α and to activate adaptive immune responses. Although IFN-α inhibits HIV-1 replication in vitro, the production of IFN-α by HIV-activated pDCs in vivo may contribute more to HIV pathogenesis than to protection. We have now shown that HIV-stimulated human pDCs allow for persistent IFN-α production upon repeated stimulation, express low levels of maturation molecules, and stimulate weak T cell responses. Persistent IFN-α production by HIV-stimulated pDCs correlated with increased levels of IRF7 and was dependent upon the autocrine IFN-α/ß receptor feedback loop. Because it has been shown that early endosomal trafficking of TLR9 agonists causes strong activation of the IFN-α pathway but weak activation of the NF-κB pathway, we sought to investigate whether early endosomal trafficking of HIV, a TLR7 agonist, leads to the IFN-α-producing phenotype we observed. We demonstrated that HIV preferentially traffics to the early endosome in human pDCs and therefore skews pDCs toward a partially matured, persistently IFN-α-secreting phenotype.


Subject(s)
Dendritic Cells/virology , HIV Infections/metabolism , HIV/metabolism , Interferon-alpha/metabolism , NF-kappa B/metabolism , Toll-Like Receptor 9/agonists , Cytokines/metabolism , Humans , Immune System , Phenotype , RNA, Messenger/metabolism , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Time Factors
15.
Blood ; 116(19): 3839-52, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20693428

ABSTRACT

Myeloid and plasmacytoid dendritic cells (DCs) are important mediators of both innate and adaptive immunity against pathogens such as HIV. During the course of HIV infection, blood DC numbers fall substantially. In the present study, we sought to determine how early in HIV infection the reduction occurs and whether the remaining DC subsets maintain functional capacity. We find that both myeloid DC and plasmacytoid DC levels decline very early during acute HIV infection. Despite the initial reduction in numbers, those DCs that remain in circulation retain their function and are able to stimulate allogeneic T-cell responses, and up-regulate maturation markers plus produce cytokines/chemokines in response to stimulation with TLR7/8 agonists. Notably, DCs from HIV-infected subjects produced significantly higher levels of cytokines/chemokines in response to stimulation with TLR7/8 agonists than DCs from uninfected controls. Further examination of gene expression profiles indicated in vivo activation, either directly or indirectly, of DCs during HIV infection. Taken together, our data demonstrate that despite the reduction in circulating DC numbers, those that remain in the blood display hyperfunctionality and implicates a possible role for DCs in promoting chronic immune activation.


Subject(s)
Dendritic Cells/immunology , HIV Infections/immunology , Blood Cell Count , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Case-Control Studies , Chemokines/biosynthesis , Chemokines/genetics , Cross-Sectional Studies , Cytokines/biosynthesis , Cytokines/genetics , Dendritic Cells/classification , Gene Expression , HIV Infections/blood , HIV Infections/genetics , HIV Infections/virology , Humans , In Vitro Techniques , Longitudinal Studies , Lymphocyte Activation , Signal Transduction , Time Factors , Toll-Like Receptors/agonists , Viral Load
16.
Immunotherapy ; 2(1): 37-56, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20473346

ABSTRACT

The use of dendritic cells (DCs) for tumor immunotherapy represents a powerful approach for harnessing the patient's own immune system to eliminate tumor cells. However, suboptimal conditions for generating potent immunostimulatory DCs, as well as the induction of tolerance and suppression mediated by the tumors and its microenvironment have contributed to limited success. Combining DC vaccines with new approaches that enhance immunogenicity and overcome the regulatory mechanisms underlying peripheral tolerance may be the key to achieving effective and durable anti-tumor immune responses that translate to better clinical outcomes.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Immunotherapy/methods , Neoplasms/therapy , Antigen-Presenting Cells/immunology , Cell Differentiation/immunology , Humans , Immune Tolerance/immunology , Neoplasms/immunology
17.
Eur J Immunol ; 40(8): 2248-58, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20455275

ABSTRACT

Priming of T cells in lymphoid tissues of HIV-infected individuals occurs in the presence of HIV-1. DC in this milieu activate T cells and disseminate HIV-1 to newly activated T cells, the outcome of which may have serious implications in the development of optimal antiviral responses. We investigated the effects of HIV-1 on DC-naïve T-cell interactions using an allogeneic in vitro system. Our data demonstrate a dramatic decrease in the primary expansion of naïve T cells when cultured with HIV-1-exposed DC. CD4(+) and CD8(+) T cells showed enhanced expression of PD-1 and TRAIL, whereas CTLA-4 expression was observed on CD4(+) T cells. It is worth noting that T cells primed in the presence of HIV-1 suppressed priming of other naïve T cells in a contact-dependent manner. We identified PD-1, CTLA-4, and TRAIL pathways as responsible for this suppresion, as blocking these negative molecules restored T-cell proliferation to a higher degree. In conclusion, the presence of HIV-1 during DC priming produced cells with inhibitory effects on T-cell activation and proliferation, i.e. suppressor T cells, a mechanism that could contribute to the enhancement of HIV-1 pathogenesis.


Subject(s)
Dendritic Cells/metabolism , HIV Infections/immunology , HIV-1/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , 2,2'-Dipyridyl/analogs & derivatives , 2,2'-Dipyridyl/chemistry , Antibodies, Blocking/pharmacology , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , CTLA-4 Antigen , Cell Communication , Cell Proliferation/drug effects , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Disulfides/chemistry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , HIV Antigens/immunology , HIV Antigens/metabolism , HIV-1/chemistry , HIV-1/pathogenicity , Humans , Lymphocyte Activation/drug effects , Programmed Cell Death 1 Receptor , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/virology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/virology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/immunology , TNF-Related Apoptosis-Inducing Ligand/metabolism
18.
PLoS One ; 4(1): e4256, 2009.
Article in English | MEDLINE | ID: mdl-19165342

ABSTRACT

BACKGROUND: The requirements for priming of HIV-specific T cell responses initially seen in infected individuals remain to be defined. Activation of T cell responses in lymph nodes requires cell-cell contact between T cells and DCs, which can give concurrent activation of T cells and HIV transmission. METHODOLOGY: The study aim was to establish whether DCs pulsed with HIV-1 could prime HIV-specific T cell responses and to characterize these responses. Both infectious and aldrithiol-2 inactivated noninfectious HIV-1 were compared to establish efficiencies in priming and the type of responses elicited. FINDINGS: Our findings show that both infectious and inactivated HIV-1 pulsed DCs can prime HIV-specific responses from naïve T cells. Responses included several CD4(+) and CD8(+) T cell epitopes shown to be recognized in vivo by acutely and chronically infected individuals and some CD4(+) T cell epitopes not identified previously. Follow up studies of acute and recent HIV infected samples revealed that these latter epitopes are among the earliest recognized in vivo, but the responses are lost rapidly, presumably through activation-induced general CD4(+) T cell depletion which renders the newly activated HIV-specific CD4(+) T cells prime targets for elimination. CONCLUSION: Our studies highlight the ability of DCs to efficiently prime naïve T cells and induce a broad repertoire of HIV-specific responses and also provide valuable insights to the pathogenesis of HIV-1 infection in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/virology , HIV Infections/blood , HIV Infections/virology , HIV-1/metabolism , 2,2'-Dipyridyl/analogs & derivatives , 2,2'-Dipyridyl/metabolism , Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Communication , Dendritic Cells/cytology , Disulfides/metabolism , Epitopes , Humans , L-Selectin/biosynthesis , Leukocyte Common Antigens/biosynthesis , Leukocytes, Mononuclear/virology , Lymphocyte Activation , Phenotype
19.
Eur J Immunol ; 37(7): 1752-63, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17534864

ABSTRACT

The outcome following HIV infection depends on the nature and durability of the HIV-specific T cell response induced initially. The activation of protective T cell responses depends upon dendritic cells (DC), antigen-presenting cells which have the capacity to process and present viral antigens. DC pulsed with aldrithiol-2-inactivated HIV and delivered in vivo were reported to induce immune responses and promote virologic control in chronically HIV-1-infected subjects. To gain an understanding of this phenomenon, we characterized the steps involved in the presentation of antigens derived from aldrithiol-2-treated vs. infectious HIV-1 by DC. Antigen presentation, on both MHC class I and II, was independent of DC-specific ICAM-3-grabbing integrin, DEC-205 and macrophage mannose receptor, C-type lectins expressed by the DC. Inhibitor studies showed that presentation on MHC class I was dependent on viral fusion in a CD4/coreceptor-dependent manner, both at the cell surface and within endosomes, and access to the classical endosomal processing pathway. MHC class II presentation of HIV-associated antigens was dependent on active endocytosis, probably receptor-mediated, and subsequent degradation of virions in acidified endosomes in the DC. Our study brings forth new facts regarding the binding, uptake, and processing of chemically inactivated virions leading to efficient antigen presentation and should aid in the design of more effective HIV vaccines.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , HIV Antigens/immunology , HIV-1/immunology , 2,2'-Dipyridyl/analogs & derivatives , 2,2'-Dipyridyl/pharmacology , Antiviral Agents/pharmacology , Disulfides/pharmacology , HIV-1/drug effects , Histocompatibility Antigens Class I , Histocompatibility Antigens Class II , Humans , Virion/drug effects , Virion/immunology
20.
J Virol ; 79(24): 15368-75, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16306608

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) evolves in vivo under selective pressure from CD8+ T-lymphocyte (CTL) responses, which are in turn determined by host and viral genetic factors, such as restricting major histocompatibility complex molecules and the available viral epitope sequences. However, CTL are derived stochastically through the random gene rearrangements to produce T-cell receptors (TCR), and the relative impact of genetic versus stochastic processes on CTL targeting of HIV and immune-driven viral evolution is unclear. Here we evaluate identical twins infected with HIV-1 as neonates from a common blood transfusion, with subsequently similar environmental exposures, thereby allowing controlled comparisons of CTL targeting and viral evolution. Seventeen years after infection, their CTL targeting of HIV-1 was remarkably similar. In contrast, their overall TCR profiles were highly dissimilar, and a dominant epitope was recognized by distinctly different TCR in each twin. Furthermore, their viral epitopes had diverged, and there was ongoing viral phylogenetic divergence between the twins between 12 and 17 years after infection. These results indicate that while CTL targeting is predominately genetically determined, stochastic influences render the interaction of HIV-1 and host immunity, and therefore viral escape and CTL efficacy, unpredictable.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , HIV-1/metabolism , T-Lymphocytes, Cytotoxic/metabolism , HIV Seropositivity/immunology , HIV Seropositivity/microbiology , HIV-1/immunology , Humans , Immunodominant Epitopes/immunology , T-Lymphocytes, Cytotoxic/immunology , Twins
SELECTION OF CITATIONS
SEARCH DETAIL
...