Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Front Immunol ; 13: 867013, 2022.
Article in English | MEDLINE | ID: mdl-35757746

ABSTRACT

Adoptive transfer of chimeric antigen receptor (CAR) T lymphocytes is a powerful technology that has revolutionized the way we conceive immunotherapy. The impressive clinical results of complete and prolonged response in refractory and relapsed diseases have shifted the landscape of treatment for hematological malignancies, particularly those of lymphoid origin, and opens up new possibilities for the treatment of solid neoplasms. However, the widening use of cell therapy is hampered by the accessibility to viral vectors that are commonly used for T cell transfection. In the era of messenger RNA (mRNA) vaccines and CRISPR/Cas (clustered regularly interspaced short palindromic repeat-CRISPR-associated) precise genome editing, novel and virus-free methods for T cell engineering are emerging as a more versatile, flexible, and sustainable alternative for next-generation CAR T cell manufacturing. Here, we discuss how the use of non-viral vectors can address some of the limitations of the viral methods of gene transfer and allow us to deliver genetic information in a stable, effective and straightforward manner. In particular, we address the main transposon systems such as Sleeping Beauty (SB) and piggyBac (PB), the utilization of mRNA, and innovative approaches of nanotechnology like Lipid-based and Polymer-based DNA nanocarriers and nanovectors. We also describe the most relevant preclinical data that have recently led to the use of non-viral gene therapy in emerging clinical trials, and the related safety and efficacy aspects. We will also provide practical considerations for future trials to enable successful and safe cell therapy with non-viral methods for CAR T cell generation.


Subject(s)
Immunotherapy, Adoptive , Receptors, Antigen, T-Cell , Gene Editing/methods , Immunotherapy, Adoptive/methods , RNA, Messenger , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes
2.
Sci Rep ; 10(1): 5134, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32198428

ABSTRACT

Approaches to deplete persistent HIV infection are needed. We investigated the combined impact of the latency reversing agent vorinostat (VOR) and AGS-004, an autologous dendritic cell immunotherapeutic, on the HIV reservoir. HIV+, stably treated participants in whom resting CD4+ T cell-associated HIV RNA (rca-RNA) increased after VOR exposure ex vivo and in vivo received 4 doses of AGS-004 every 3 weeks, followed by VOR every 72 hours for 30 days, and then the cycle repeated. Change in VOR-responsive host gene expression, HIV-specific T cell responses, low-level HIV viremia, rca-RNA, and the frequency of resting CD4+ T-cell infection (RCI) was measured at baseline and after each cycle. No serious treatment-related adverse events were observed among five participants. As predicted, VOR-responsive host genes responded uniformly to VOR dosing. Following cycles of AGS-004 and VOR, rca-RNA decreased significantly in only two participants, with a significant decrease in SCA observed in one of these participants. However, unlike other cohorts dosed with AGS-004, no uniform increase in HIV-specific immune responses following vaccination was observed. Finally, no reproducible decline of RCI, defined as a decrease of >50%, was observed. AGS-004 and VOR were safe and well-tolerated, but no substantial impact on RCI was measured. In contrast to previous clinical data, AGS-004 did not induce HIV-specific immune responses greater than those measured at baseline. More efficacious antiviral immune interventions, perhaps paired with more effective latency reversal, must be developed to clear persistent HIV infection.


Subject(s)
Dendritic Cells/transplantation , HIV Infections/therapy , HIV-1/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Immunotherapy, Adoptive/methods , Vorinostat/therapeutic use , Adult , CD4-Positive T-Lymphocytes/immunology , Humans , Immunologic Memory/immunology , Male , Middle Aged , T-Lymphocytes, Cytotoxic/immunology , Translational Research, Biomedical , Treatment Outcome , Vaccination
3.
Clin Cancer Res ; 26(10): 2327-2336, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32034074

ABSTRACT

PURPOSE: Rocapuldencel-T is an autologous immunotherapy prepared from mature monocyte-derived dendritic cells (DC), coelectroporated with amplified tumor RNA plus CD40L RNA. This pivotal phase III trial was initiated to investigate the safety and efficacy of a combination therapy dosing regimen of Rocapuldencel-T plus sunitinib in patients with metastatic renal cell carcinoma (mRCC). PATIENTS AND METHODS: Patients received either Rocapuldencel-T plus standard of care (SOC) or SOC treatment alone. The primary objective compared overall survival (OS) between groups. Secondary objectives included safety assessments, progression-free survival (PFS), and tumor responses based on RECIST 1.1 criteria. Exploratory analyses included immunologic assessments and correlates with OS. RESULTS: Between 2013 and 2016, 462 patients were randomized 2:1, 307 to the combination group and 155 to the SOC group. Median OS in the combination group was 27.7 months [95% confidence interval (CI) 23.0-35.9] and 32.4 months (95% CI, 22.5-) in the SOC group HR of 1.10 (95% CI, 0.83-1.40). PFS was 6.0 months and 7.83 months for the combination and SOC groups, respectively [HR = 1.15 (95% CI, 0.92-1.44)]. The ORR was 42.7% (95% CI, 37.1-48.4) for the combination group and 39.4% (95% CI, 31.6-47.5) for the SOC group. Median follow up was 29 months (0.4-47.7 months). On the basis of the lack of clinical efficacy, the ADAPT trial was terminated on February 17, 2017. Immune responses were detected in 70% of patients treated with Rocapuldencel-T, and the magnitude of the immune response positively correlated with OS. In addition, we report the survival-predictive value of measuring IL-12 produced by the DC vaccine and the observation that high baseline numbers of T regulatory cells are associated with improved outcomes in DC-treated patients, but are associated with poor outcomes in patients receiving SOC treatment. No serious adverse events attributed to the study medication have been reported to date. CONCLUSIONS: Rocapuldencel-T did not improve OS in patients treated with combination therapy, although the induced immune response correlated with OS. Moreover, we identified two potential survival-predictive biomarkers for patients receiving DC based immunotherapy, IL-12 produced by the DC vaccine and higher numbers of T regulatory cells present in the peripheral blood of patients with advanced RCC.


Subject(s)
Antigens, Neoplasm/immunology , Carcinoma, Renal Cell/therapy , Dendritic Cells/transplantation , Immunotherapy/methods , Kidney Neoplasms/therapy , Sunitinib/therapeutic use , Antigen Presentation/immunology , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Combined Modality Therapy , Dendritic Cells/immunology , Female , Follow-Up Studies , Humans , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Lymphatic Metastasis , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate , T-Lymphocytes, Regulatory/immunology
4.
AIDS Res Hum Retroviruses ; 34(1): 111-122, 2018 01.
Article in English | MEDLINE | ID: mdl-28636433

ABSTRACT

AGS-004 consists of matured autologous dendritic cells co-electroporated with in vitro transcribed RNA encoding autologous HIV antigens. In an open-label, single arm sub-study of AGS-004-003, AGS-004 was administered monthly to suppressed participants who started antiretroviral therapy (ART) during acute HIV infection. HIV-1 specific T cell responses were measured by multicolor flow cytometry after 3-4 doses. The frequency of resting CD4+ T-cell infection (RCI) was measured by quantitative viral outgrowth assay. Participants demonstrating increased immune response postvaccination were eligible for analytic treatment interruption (ATI). AGS-004 induced a positive immune response defined as ≥2-fold increase from baseline in the number of multifunctional HIV-1 specific CD28+/CD45RA- CD8+ effector/memory cytoxic T-lymphocytes (CTLs) in all six participants. All participants underwent ATI with rebound viremia at a median of 29 days. Immune correlates between time to viral rebound and the induction of effector CTLs were determined. Baseline RCI was low in most participants (0.043-0.767 IUPM). One participant had a >2-fold decrease (0.179-0.067 infectious units per million [IUPM]) in RCI at week 10. One participant with the lowest RCI had the longest ATI. AGS-004 dendritic cell administration increased multifunctional HIV-specific CD28+/CD45RA- CD8+ memory T cell responses in all participants, but did not permit sustained ART interruption. However, greater expansion of CD28-/CCR7-/CD45RA- CD8+ effector T cell responses correlated with a longer time to viral rebound. AGS-004 may be a useful tool to augment immune responses in the setting of latency reversal and eradication strategies.


Subject(s)
Dendritic Cells/immunology , HIV Infections/immunology , HIV Infections/therapy , Immunogenicity, Vaccine , Immunotherapy/methods , Acute Disease , Adolescent , Adult , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , HIV-1 , Humans , Male , Middle Aged , RNA, Viral , Viral Load , Viremia , Young Adult
5.
J Immunol ; 198(6): 2286-2301, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28193829

ABSTRACT

The transmembrane protein CD83, expressed on APCs, B cells, and T cells, can be expressed as a soluble form generated by alternative splice variants and/or by shedding. Soluble CD83 (sCD83) was shown to be involved in negatively regulating the immune response. sCD83 inhibits T cell proliferation in vitro, supports allograft survival in vivo, prevents corneal transplant rejection, and attenuates the progression and severity of autoimmune diseases and experimental colitis. Although sCD83 binds to human PBMCs, the specific molecules that bind sCD83 have not been identified. In this article, we identify myeloid differentiation factor-2 (MD-2), the coreceptor within the TLR4/MD-2 receptor complex, as the high-affinity sCD83 binding partner. TLR4/MD-2 mediates proinflammatory signal delivery following recognition of bacterial LPSs. However, altering TLR4 signaling can attenuate the proinflammatory cascade, leading to LPS tolerance. Our data show that binding of sCD83 to MD-2 alters this signaling cascade by rapidly degrading IL-1R-associated kinase-1, leading to induction of the anti-inflammatory mediators IDO, IL-10, and PGE2 in a COX-2-dependent manner. sCD83 inhibited T cell proliferation, blocked IL-2 secretion, and rendered T cells unresponsive to further downstream differentiation signals mediated by IL-2. Therefore, we propose the tolerogenic mechanism of action of sCD83 to be dependent on initial interaction with APCs, altering early cytokine signal pathways and leading to T cell unresponsiveness.


Subject(s)
Antigens, CD/metabolism , Immunoglobulins/metabolism , Membrane Glycoproteins/metabolism , Monocytes/immunology , T-Lymphocytes/immunology , Cell Proliferation , Cells, Cultured , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-10/metabolism , Interleukin-2/metabolism , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/immunology , Lymphocyte Activation , Lymphocyte Antigen 96/metabolism , Protein Binding , Signal Transduction , Toll-Like Receptor 4/metabolism , CD83 Antigen
6.
J Acquir Immune Defic Syndr ; 72(1): 31-8, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26751016

ABSTRACT

BACKGROUND: The genomic heterogeneity of HIV-1 impedes the ability of consensus sequences in vaccines to elicit effective antiviral immune responses. AGS-004 amplifies translation-competent RNA molecules encoding for Gag, Rev, Vpr, and Nef from the patient's autologous virus and loads them into dendritic cells. METHODS: This phase IIB, multicenter, 2:1 randomized, double-blind, placebo-controlled study enrolled 54 HIV-1-infected patients on antiretroviral therapy with viral loads (VLs) <50 copies per milliliter, current CD4 T-cell counts >450 cells per cubic millimeter, and nadir counts >200 cells per cubic millimeter, to receive intradermal injections of study product into the axillary lymph node region every 4 weeks. At week 16, a 12-week analytical treatment interruption (ATI) was undertaken. RESULTS: There was no difference in the end-of-ATI VL (average of values from weeks 11 and 12) between the 2 arms of the study [4.39 (4.17, 4.69) vs. 4.47 (3.76, 4.64) log10 HIV-1 RNA; P = 0.73]. Between arms, no change between pre-antiretroviral therapy VL and the end-of-ATI VL [-0.06 (0.24, -0.32) vs. -0.17 (0.17, -0.32) log10 HIV-1 RNA; P = 0.43] was observed. When interferon-γ, interleukin-2, tumor necrosis factor α, CD107a, and granzyme b expressions were measured by multicolor flow cytometry, a greater percentage of AGS-004 than of placebo recipients had multifunctional cytotoxic T-lymphocyte responses induced in the CD28+/CD45RA-CD8 effector/memory T-cell population to dendritic cells electroporated with autologous antigens. Adverse events consisted of transient, mild (grade 1) local injection site reactions. CONCLUSIONS: Despite the induction of HIV-specific effector/memory CD8 T-cell responses, no antiviral effect was seen after the administration of AGS-004 when compared with placebo.


Subject(s)
Dendritic Cells/immunology , HIV Infections/therapy , HIV-1/immunology , Immunotherapy/methods , RNA, Viral/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Adolescent , Adult , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Double-Blind Method , Female , Granzymes/biosynthesis , HIV-1/genetics , Humans , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lysosomal-Associated Membrane Protein 1/biosynthesis , Male , Middle Aged , Placebos/therapeutic use , Tumor Necrosis Factor-alpha/biosynthesis , Viral Load , Young Adult , gag Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/genetics , rev Gene Products, Human Immunodeficiency Virus/genetics , vpr Gene Products, Human Immunodeficiency Virus/genetics
7.
J Immunother Cancer ; 3: 14, 2015.
Article in English | MEDLINE | ID: mdl-25901286

ABSTRACT

BACKGROUND: AGS-003 is an autologous immunotherapy prepared from fully matured and optimized monocyte-derived dendritic cells, which are co-electroporated with amplified tumor RNA plus synthetic CD40L RNA. AGS-003 was evaluated in combination with sunitinib in an open label phase 2 study in intermediate and poor risk, treatment naïve patients with metastatic clear cell renal cell carcinoma (mRCC). METHODS: Twenty-one intermediate and poor risk patients were treated continuously with sunitinib (4 weeks on, 2 weeks off per 6 week cycle). After completion of the first cycle of sunitinib, patients were treated with AGS-003 every 3 weeks for 5 doses, then every 12 weeks until progression or end of study. The primary endpoint was to determine the complete response rate. Secondary endpoints included clinical benefit, safety, progression free survival (PFS) and overall survival (OS). Immunologic response was also monitored. RESULTS: Thirteen patients (62%) experienced clinical benefit (9 partial responses, 4 with stable disease); however there were no complete responses in this group of intermediate and poor risk mRCC patients and enrollment was terminated early. Median PFS from registration was 11.2 months (95% CI 6.0, 19.4) and the median OS from registration was 30.2 months (95% CI 9.4, 57.1) for all patients. Seven (33%) patients survived for at least 4.5 years, while five (24%) survived for more than 5 years, including 2 patients who remain progression-free with durable responses for more than 5 years at the time of this report. AGS-003 was well tolerated with only mild injection-site reactions. The most common adverse events were related to expected toxicity from sunitinib therapy. In patients who had sequential samples available for immune monitoring, the magnitude of the increase in the absolute number of CD8(+) CD28(+) CD45RA(-) effector/memory T cells (CTLs) after 5 doses of AGS-003 relative to baseline, correlated with overall survival. CONCLUSIONS: AGS-003 in combination with sunitinib was well tolerated and yielded supportive immunologic responses coupled with extension of median and long-term survival in an unselected, intermediate and poor risk prognosis mRCC population. CLINICAL TRIAL REGISTRY: #NCT00678119.

8.
J Acquir Immune Defic Syndr ; 64(4): 345-50, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24025338

ABSTRACT

We previously reported that a combination of antiretroviral therapy with 4 monthly injections of each patient's own autologous dendritic cells (AGS-004) electroporated with CD40 ligand and with HIV RNA antigens obtained from each patient's own pre-antiretroviral therapy plasma induced HIV-specific CD8 T-cell responses in 10 patients. To assess other AGS-004-induced immune changes, we evaluated the modifications in B- and T-cell subsets and the level of immune activation in these patients. The proportion of Bm1 naive cells was increased along with an augmentation of the proliferation marker Ki67. Memory B-cell frequency, CD4 and CD8 T-cell subsets, regulatory T-cell frequency, and CD38/HLA-DR/PD-1 T-cell activation levels remained unchanged after AGS-004 dendritic cell immunotherapy.


Subject(s)
B-Lymphocyte Subsets/physiology , Dendritic Cells/physiology , HIV Infections/therapy , Immunotherapy/methods , T-Lymphocyte Subsets/physiology , CD4 Lymphocyte Count , HIV Infections/immunology , Humans , RNA, Viral , Viral Load , Virus Replication
9.
J Immunol ; 191(4): 1965-75, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23851696

ABSTRACT

Modulation of immune responses is one of the main research aims in transplant immunology. In this study, we investigate the local immunomodulatory properties of soluble CD83 (sCD83) at the graft-host interface using the high-risk corneal transplantation model. In this model, which mimics the inflammatory status and the preexisting vascularization of high-risk patients undergoing corneal transplantation, allogeneic donor corneas are transplanted onto sCD83-treated recipient animals. This model allows the direct and precise application of the immune modulator at the transplantation side. Interestingly, sCD83 was able to prolong graft survival after systemic application as well as after topical application, which is therapeutically more relevant. The therapeutic effect was accompanied by an increase in the frequency of regulatory T cells and was mediated by the immune-regulatory enzyme IDO and TGF-ß. In vitro, sCD83 induced long-term IDO expression in both conventional and plasmacytoid dendritic cells via autocrine or paracrine production of TGF-ß, a cytokine previously shown to be an essential mediator of IDO-dependent, long-term tolerance. These findings open new treatment avenues for local immune modulation after organ and tissue transplantation.


Subject(s)
Antigens, CD/therapeutic use , Corneal Transplantation , Graft Enhancement, Immunologic , Immunoglobulins/therapeutic use , Immunologic Factors/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Membrane Glycoproteins/therapeutic use , T-Lymphocytes, Regulatory/drug effects , Transplantation Tolerance/drug effects , Administration, Ophthalmic , Allografts , Animals , Antigens, CD/administration & dosage , Antigens, CD/immunology , Bone Marrow Cells/immunology , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , Female , Forkhead Transcription Factors/analysis , Graft Survival , Immunoglobulins/administration & dosage , Immunoglobulins/immunology , Immunologic Factors/administration & dosage , Immunologic Factors/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Injections, Intraperitoneal , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Premedication , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Solubility , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/administration & dosage , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta/therapeutic use , CD83 Antigen
10.
Mol Ther Nucleic Acids ; 2: e91, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23653155

ABSTRACT

Dendritic cells (DCs) transfected with total amplified tumor cell RNA have the potential to induce broad antitumor immune responses. However, analytical methods required for quantitatively assessing the integrity, fidelity, and functionality of the amplified RNA are lacking. We have developed a series of assays including gel electrophoresis, northern blot, capping efficiency, and microarray analysis to determine integrity and fidelity and a model system to assess functionality after transfection into human DCs. We employed these tools to demonstrate that modifications to our previously reported total cellular RNA amplification process including the use of the Fast Start High Fidelity (FSHF) PCR enzyme, T7 Powerswitch primer, post-transcriptional capping and incorporation of a type 1 cap result in amplification of longer transcripts, greater translational competence, and a higher fidelity representation of the starting total RNA population. To study the properties of amplified RNA after transfection into human DCs, we measured protein expression levels of defined antigens coamplified with the starting total RNA populations and measured antigen-specific T cell expansion in autologous DC-T cell co-cultured in vitro. We conclude from these analyses that the improved RNA amplification process results in superior protein expression levels and a greater capacity of the transfected DCs to induce multifunctional antigen-specific memory T cells.Molecular Therapy-Nucleic Acids (2013) 2, e91; doi:10.1038/mtna.2013.18; published online 7 May 2013.

11.
J Pharm Biomed Anal ; 70: 657-63, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22703839

ABSTRACT

Ribonucleic acid (RNA) is gaining utility as a key component of immunotherapeutics to transiently express antigens or to modulate endogenous gene expression for clinical applications. As a key ancillary component of clinical grade products, RNA requires a robust method for quality control. Here we evaluated the microfluidics based platform and slab electrophoresis for determination of integrity, concentration and size of four in vitro-transcribed RNA products with sizes of 1611, 808, 475 and 290 nucleotides (nts). Our data demonstrate that the Bioanalyzer can determine both size and integrity of the RNA, but the analysis suffers from a strong well position effect. For the RNAs tested, the integrity values obtained by the Bioanalyzer demonstrate a reverse correlation with the size of the molecule and are lower than those obtained using slab electrophoresis. Agarose gel electrophoresis produced the information on size of the RNA molecule with good precision, accuracy and reproducibility. We highlight observations which need to be taken into account when developing and qualifying a method of choice for assessment of in vitro-transcribed RNA using either approach.


Subject(s)
Biotechnology/methods , Electrophoresis, Agar Gel , Electrophoresis, Microchip , Microfluidic Analytical Techniques , RNA/biosynthesis , Transcription, Genetic , Biotechnology/standards , Electrophoresis, Agar Gel/standards , Electrophoresis, Microchip/standards , Microfluidic Analytical Techniques/standards , Quality Control , RNA/analysis , RNA/standards , RNA/therapeutic use , Reproducibility of Results , Ribonucleotides/analysis , Spectrophotometry
12.
J Immunother ; 34(1): 45-57, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21150712

ABSTRACT

Electroporation of mature dendritic cells (DC) with RNA-encoding CD40L greatly enhances the production of interleukin (IL)-12, a proinflammatory cytokine necessary for the induction of T-cell immunity. Results presented herein reveal a correlation between the priming of CD28(+) antigen-reactive effector memory cytotoxic T lymphocytes (CTL) displaying 3 or 4 simultaneous effector functions and the quantity of IL-12 produced by postmaturation electroporation-CD40L DC. By using multiparameter flow cytometry, the quantities of IL-12 needed to prime naive antigen-reactive T cells to simultaneously produce interferon-γ and tumor necrosis factor-α in the presence or absence of IL-2 secretion in conjunction with lytic activity defined by CD107a expression can be used to determine the overall potency of a DC product. In the presence of IL-12, CTL differentiation toward lytic function is not accompanied by a reduction in the secretion of interferon-γ and tumor necrosis factor-α. Therefore, by measuring the availability of IL-12 one can predict the potency of a DC immunotherapeutics in relation to its ability to drive distinct effector memory CTL subsets with multifunctional activities.


Subject(s)
CD40 Ligand/genetics , Dendritic Cells/metabolism , Electroporation , Interleukin-12/metabolism , RNA/genetics , T-Lymphocytes, Cytotoxic/immunology , CD28 Antigens/biosynthesis , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Interleukin-2/immunology , Interleukin-2/metabolism , Lymphocyte Activation , Lysosomal-Associated Membrane Protein 1/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis
13.
Mol Ther ; 18(12): 2112-20, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20628358

ABSTRACT

Bone marrow-derived dendritic cells (DCs) are cells of the immune system that have been used as a tool to boost, modulate, or dampen immune responses. In the context of autoimmunity, DCs can be modified to express immunoregulatory products encoded by transgenes, and used therapeutically in adoptive cellular therapy. DCs that were lentivirally transduced (lt) to express interleukin 4 (IL-4) can significantly delay or prevent the onset of autoimmune diabetes in nonobese diabetic (NOD) mice. However, modifying cells using viral vectors carries the dual risk of oncogenicity or immunogenicity. This study demonstrates that NOD DCs, electroporated with "translationally enhanced" IL-4 mRNA (eDC/IL-4), can be equally efficient therapeutically, despite the reduced amount and shorter duration of IL-4 secretion. Moreover, a single injection of eDC/IL-4 in NOD mice shortly after the onset of hyperglycemia was able to maintain stable glycemia for up to several months in a significant fraction of treated mice. Treatment with eDC/IL-4 boosted regulatory T (Tregs) cell functions and modulated T helper responses to reduce pathogenicity. Thus, treatment with DCs, electroporated with modified IL-4 mRNA to express IL-4 for up to 24 hours, constitutes a viable cellular therapy approach for the regulation of autoimmune diabetes, as a preferred alternative to the use of viral vectors.


Subject(s)
Dendritic Cells/immunology , Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 1/therapy , Interleukin-4/therapeutic use , Animals , Dendritic Cells/transplantation , Electroporation , Interleukin-4/administration & dosage , Mice , Mice, Inbred NOD , RNA, Messenger/genetics
14.
Clin Immunol ; 134(2): 140-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19889582

ABSTRACT

Immunogenicity, manufacturing feasibility, and safety of a novel, autologous dendritic cell (DC)-based immunotherapy (AGS-004) was evaluated in ten human immunodeficiency virus type 1 (HIV-1)-infected adults successfully treated with antiretroviral therapy (ART). Personalized AGS-004 was produced from autologous monocyte-derived DCs electroporated with RNA encoding CD40L and HIV antigens (Gag, Vpr, Rev, and Nef) derived from each subjects' pre-ART plasma. Patients received monthly injections of AGS-004 in combination with ART. AGS-004 was produced within a mean of 6 weeks and yielded 4-12 doses/subject Full or partial HIV-specific proliferative immune responses occurred in 7 of 9 evaluable subjects. Responses were specific for the AGS-004 presented HIV antigens and preferentially targeted CD8(+) T cells. Mild adverse events included flu-like symptoms, fatigue, and injection site reactions. No evidence of autoimmunity, changes in viral load, or significant changes in absolute CD4(+) and CD8(+) T cell counts were observed. This pilot study supports the further clinical investigation of AGS-004.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/transplantation , HIV Infections/therapy , Immunotherapy/methods , RNA, Viral/immunology , Adult , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Count , Electroporation , HIV/immunology , HIV Infections/immunology , Humans , Male , Middle Aged , Pilot Projects , gag Gene Products, Human Immunodeficiency Virus/immunology , nef Gene Products, Human Immunodeficiency Virus/immunology , rev Gene Products, Human Immunodeficiency Virus/immunology , vpr Gene Products, Human Immunodeficiency Virus/immunology
15.
PLoS One ; 4(6): e5853, 2009 Jun 10.
Article in English | MEDLINE | ID: mdl-19516896

ABSTRACT

BACKGROUND: A hallmark of AIDS progression is a switch of cytokines from Th1 to Th2 in the plasma of patients. IL-12, a critical Th1 cytokine secreted by antigen presenting cells (APCs) is suppressed by Vpr, implicating it as an important virulence factor. We hypothesize that Vpr protein packaged in the virion may be required for disabling APCs of the first infected mucosal tissues. Consistent with this idea are reports that defects in the C-terminus of Vpr are associated with long-term non-progression. PRINCIPAL FINDINGS: Vpr RNA amplified from various sources was electroporated into monocyte-derived DC and IL-12 levels in supernatants were analyzed. The analysis of previously reported C-terminal Vpr mutations demonstrate that they do not alleviate the block of IL-12 secretion. However, a novel single conservative amino acid substitution, R90K, reverses the IL-12 suppression. Analysis of 1226 Vpr protein sequences demonstrated arginine (R) present at position 90 in 98.8%, with other substitutions at low frequency. Furthermore, none of sequences report lysine (K) in position 90. Vpr clones harboring the reported substitutions in position 90 were studied for their ability to suppress IL-12. Our data demonstrates that none of tested substitutions other than K relieve IL-12 suppression. This suggests a natural selection for sequences which suppress IL-12 secretion by DC and against mutations which relieve such suppression. Further analyses demonstrated that the R90K, as well as deletion of the C-terminus, directs the Vpr protein for rapid degradation. CONCLUSION: This study supports Vpr as an HIV virulence factor during HIV infection and for the first time provides a link between evolutionary conservation of Vpr and its ability to suppress IL-12 secretion by DC. DC activated in the presence of Vpr would be defective in the production of IL-12, thus contributing to the prevailing Th2 cytokine profile associated with progressive HIV disease. These findings should be considered in the design of future immunotherapies that incorporate Vpr as an antigen.


Subject(s)
Gene Products, vpr/metabolism , HIV Infections/metabolism , Immunosuppressive Agents/pharmacology , Interleukin-12/metabolism , Mucous Membrane/virology , vpr Gene Products, Human Immunodeficiency Virus/chemistry , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Dendritic Cells/metabolism , Humans , Mucous Membrane/metabolism , Mutation , Plasmids/metabolism , Protein Structure, Tertiary , RNA, Viral/metabolism , Transcription, Genetic , Ubiquitin/metabolism , Virulence Factors
16.
J Immunol ; 181(8): 5296-305, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832685

ABSTRACT

Dendritic cell (DC)-based immunotherapeutics must induce robust CTL capable of killing tumor or virally infected cells in vivo. In this study, we show that RNA electroporated post maturation and coelectroporated with CD40L mRNA (post maturation electroporation (PME)-CD40L DC) generate high-avidity CTL in vitro that lyse naturally processed and presented tumor Ag. Unlike cytokine mixture-matured DC which induce predominantly nonproliferative effector memory CD45RA(+) CTL, PME-CD40L DC prime a novel subset of Ag-specific CTL that can be expanded to large numbers upon sequential DC stimulation in vitro. We have defined these cells as rapidly expanding high-avidity (REHA) CTL based on: 1) the maintenance of CD28 expression, 2) production of high levels of IFN-gamma and IL-2 in response to Ag, and 3) the demonstration of high-avidity TCR that exhibit strong cytolytic activity toward limiting amounts of native Ag. We demonstrate that induction of REHA CTL is dependent at least in part on the production of IL-12. Interestingly, neutralization of IL-12 did not effect cytolytic activity of REHA CTL when Ag is not limiting, but did result in lower TCR avidity of Ag-reactive CTL. These results suggest that PME-CD40L DC are uniquely capable of delivering the complex array of signals needed to generate stable CD28(+) REHA CTL, which if generated in vivo may have significant clinical benefit for the treatment of infectious disease and cancer.


Subject(s)
Antigens, Neoplasm/immunology , CD40 Ligand/immunology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/genetics , CD28 Antigens/genetics , CD28 Antigens/immunology , CD40 Ligand/genetics , Cell Line, Tumor , Dendritic Cells/cytology , Electroporation/methods , Gene Expression Regulation/genetics , Humans , Immunologic Memory/genetics , Immunotherapy/methods , Infections/genetics , Infections/immunology , Infections/therapy , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , RNA/genetics , RNA/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/cytology
17.
BMC Mol Biol ; 9: 90, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18928538

ABSTRACT

BACKGROUND: RNA transfection into dendritic cells (DCs) is widely used to achieve antigen expression as well as to modify DC properties. CD40L is expressed by activated T cells and interacts with CD40 receptors expressed on the surface of the DCs leading to Th1 polarization. Previous studies demonstrated that ectopic CD40L expression via DNA transfection into DCs can activate the CD40 receptor signal transduction cascade. In contrast to previous reports, this study demonstrates that the same effect can be achieved when RNA encoding CD40L is electroporated into DCs as evidenced by secretion of IL-12. To achieve higher levels of IL-12 secretion, a systematic approach involving modification of coding and noncoding regions was implemented to optimize protein expression in the DCs for the purpose of increasing IL-12 secretion. RESULTS: Site-directed mutagenesis of each of the first five in-frame methionine codons in the CD40L coding sequence demonstrated that DCs expressing a truncated CD40L protein initiated from the second methionine codon secreted the highest levels of IL-12. In addition, a post-transcriptional method of capping was utilized for final modification of the CD40L RNA. This method enzymatically creates a type I cap structure identical to that found in most eukaryotic mRNAs, in contrast to the type 0 cap incorporated using the conventional co-transcriptional capping reaction. CONCLUSION: The combination of knocking out the first initiation methionine and post-transcriptional capping of the CD40L RNA allowed for approximately a one log increase in IL-12 levels by the transfected DCs. We believe this is a first report describing improved protein expression of post-transcriptionally capped RNA in DCs. The post-transcriptional capping which allows generation of a type I cap may have broad utility for optimization of protein expression from RNA in DCs and other cell types.


Subject(s)
CD40 Ligand/genetics , Dendritic Cells/metabolism , Interleukin-12/biosynthesis , RNA Processing, Post-Transcriptional , 5' Untranslated Regions/chemistry , Amino Acid Sequence , Base Sequence , CD40 Antigens/physiology , CD40 Ligand/biosynthesis , Cells, Cultured , Codon, Initiator/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , RNA Caps/metabolism , Up-Regulation
18.
J Immunother ; 31(8): 731-41, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18779746

ABSTRACT

Dendritic cells (DC) for the immunotherapy of cancer and infectious disease require the appropriate maturation and activation signals to effectively present antigen to drive a proinflammatory response. Here we present a comparison of 4 different maturation protocols for antigen-encoded mRNA electroporated DC. Two protocols rely on cytokine-induced maturation given either preelectroporation or postelectroporation. In addition to the cytokine treatment, 2 further maturation protocols use coelectroporation of CD40L mRNA, with antigen-encoding RNA, to deliver CD40 signals. There were no significant differences in expression of costimulatory molecules such as CD80, CD83, and CD86 or the levels of expression of major histocompatibility complexes. However, results indicate that delivery of an inflammatory signal that includes interferon-gamma before the CD40 signal results in high levels of expression of interleukin-12 that was not seen in the absence of CD40L mRNA. All 4 preparations could induce expansion of primary MART-1-specific CD8+ T cells from healthy donors in vitro, but only the 2 processes receiving CD40L could induce interferon-gamma expression by those responder cells. Only DC electroporated with CD40L RNA after delivery of the inflammatory signal (PME-CD40L DC), could drive the long-term expansion of MART-1-reactive cells that displayed a CD28+/CD45RA- effector/memory phenotype with strong cytolytic activity.


Subject(s)
CD40 Antigens/immunology , CD40 Ligand/genetics , Cytokines/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , CD40 Antigens/metabolism , Cell Culture Techniques , Cytokines/metabolism , Dendritic Cells/transplantation , Electroporation , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-12/immunology , Interleukin-12/metabolism , MART-1 Antigen , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , RNA, Messenger/immunology , RNA, Messenger/metabolism , T-Lymphocytes, Cytotoxic/metabolism
19.
PLoS One ; 3(1): e1489, 2008 Jan 30.
Article in English | MEDLINE | ID: mdl-18231576

ABSTRACT

BACKGROUND: Effective therapy for HIV-infected individuals remains an unmet medical need. Promising clinical trials with dendritic cell (DC)-based immunotherapy consisting of autologous DC loaded with autologous virus have been reported, however, these approaches depend on large numbers of HIV virions to generate sufficient doses for even limited treatment regimens. METHODOLOGY/PRINCIPAL FINDINGS: The present study describes a novel approach for RT-PCR amplification of HIV antigens. Previously, RT-PCR amplification of autologous viral sequences has been confounded by the high mutation rate of the virus which results in unreliable primer-template binding. To resolve this problem we developed a multiplex RT-PCR strategy that allows reliable strain-independent amplification of highly polymorphic target antigens from any patient and requires neither viral sequence data nor custom-designed PCR primers for each individual. We demonstrate the application of our RT-PCR process to amplify translationally-competent RNA encoding regions of Gag, Vpr, Rev and Nef. The products amplified using this method represent a complex mixture of autologous antigens encoded by viral quasispecies. We further demonstrate that DCs electroporated with in vitro-transcribed HIV RNAs are capable of stimulating poly-antigen-specific CD8+ T cell responses in vitro. CONCLUSION/SIGNIFICANCE: This study describes a strategy to overcome patient to patient viral diversity enabling strain-independent RT-PCR amplification of RNAs encoding sequence divergent quasispecies of Gag, Vpr, Rev and Nef from small volumes of infectious plasma. The approach allows creation of a completely autologous therapy that does not require advance knowledge of the HIV genomic sequences, does not have yield limitations and has no intact virus in the final product. The simultaneous use of autologous viral antigens and DCs may provoke broad patient-specific immune responses that could potentially induce effective control of viral loads in the absence of conventional antiretroviral drug therapy.


Subject(s)
Dendritic Cells/immunology , Genes, Viral , HIV Infections/therapy , HIV/genetics , Immunotherapy , Reverse Transcriptase Polymerase Chain Reaction/methods , Base Sequence , DNA Primers , Humans
20.
Biochim Biophys Acta ; 1724(1-2): 127-36, 2005 Jun 20.
Article in English | MEDLINE | ID: mdl-15866517

ABSTRACT

Use of antigen encoding RNA transfected Dendritic cells in the field of cancer immunotherapy has been well established. The use of RNA overcomes limitations inherent to other autologous DC-based vaccines as it does not require specific HLA haplotypes, identification and characterization of antigens, and captures the broadest antigen repertoire. RNA offers yet another advantage-it could be amplified minimizing the requirement of tumor mass for autologous vaccine production, and will afford the opportunity to treat patients with minimal tumor burden. The original procedure described for RNA amplification resulted in a proportion of RNA transcribed in the antisense orientation. This study also demonstrates that the presence of double-stranded RNA correlates with the presence of antisense RNA. Alternative design of oligonucleotides that removes sequence redundancy eliminates the formation of both antisense and double-stranded RNA species. We provide further evidence that amplified RNA containing antisense and double-stranded RNA species results in lower recovery of DCs post-transfection and maturation, presumably through sequence-specific gene silencing. The removal of the double-stranded species from amplified RNA results in higher recovery of mature autologous amplified RNA transfected dendritic cells. Higher DC yield will allow for reduction of cost of vaccine manufacturing and prolonged treatment of a patient.


Subject(s)
Antigens, Neoplasm/genetics , Cancer Vaccines/genetics , Dendritic Cells/immunology , Polymerase Chain Reaction/methods , RNA, Neoplasm/genetics , Cancer Vaccines/immunology , Humans , Oligonucleotide Array Sequence Analysis , RNA, Antisense/chemistry , RNA, Antisense/genetics , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...