Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Cancer Immunol Immunother ; 70(11): 3081-3091, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33751208

ABSTRACT

The nanoparticle complex of cholesteryl pullulan (CHP) and NY-ESO-1 antigen protein (CHP-NY-ESO-1) presents multiple epitope peptides to MHC class I and II pathways, leading to CD8+ and CD4+ T cell responses. Poly-ICLC is a synthetic, double-stranded RNA, an agonist of toll-like receptor (TLR)-3, and a cytoplasmic receptor of melanoma differentiation-associated gene (MDA)-5. It should be a suitable immune adjuvant of cancer vaccine to overcome the inhibitory tumor microenvironment. We conducted a phase 1 clinical trial of CHP-NY-ESO-1 with poly-ICLC in patients with advanced or recurrent esophageal cancer. CHP-NY-ESO-1/poly-ICLC (µg/mg) was administered at a dose of 200/0.5 or 200/1.0 (cohorts 1 and 2, respectively) every 2 weeks for a total of six doses. The primary endpoints were safety and immune response. The secondary endpoint was tumor response. In total, 16 patients were enrolled, and six patients in each cohort completed the trial. The most common adverse event (AE) was injection site skin reaction (86.7%). No grade 3 or higher drug-related AEs were observed. No tumor responses were observed, and three patients (30%) had stable disease. The immune response was comparable between the two cohorts, and all patients (100%) achieved antibody responses with a median of 2.5 vaccinations. Comparing CHP-NY-ESO-1 alone to the poly-ICLC combination, all patients in both groups exhibited antibody responses, but the titers were higher in the combination group. In a mouse model, adding anti-PD-1 antibody to the combination of CHP-NY-ESO-1/poly-ICLC suppressed the growth of NY-ESO-1-expressing tumors. Combining the vaccine with PD-1 blockade holds promise in human trials.


Subject(s)
Antigens, Neoplasm/therapeutic use , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Carboxymethylcellulose Sodium/analogs & derivatives , Esophageal Neoplasms/drug therapy , Glucans/therapeutic use , Membrane Proteins/therapeutic use , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Adjuvants, Immunologic/therapeutic use , Aged , Aged, 80 and over , Animals , Antigens, Neoplasm/immunology , Carboxymethylcellulose Sodium/therapeutic use , Esophageal Neoplasms/immunology , Female , Glucans/immunology , Humans , Interferon Inducers/immunology , Interferon Inducers/therapeutic use , Male , Membrane Proteins/immunology , Mice , Middle Aged , Nanoparticles , Poly I-C/immunology , Polylysine/immunology , Polylysine/therapeutic use
2.
Semin Immunol ; 49: 101414, 2020 06.
Article in English | MEDLINE | ID: mdl-33011064

ABSTRACT

Immunotherapies have become the first line of treatment for many cancer types. Unfortunately, only a small fraction of patients benefits from these therapies. This low rate of success can be attributed to 3 main barriers: 1) low frequency of anti-tumor specific T cells; 2) lack of infiltration of the anti-tumor specific T cells into the tumor parenchyma and 3) accumulation of highly suppressive cells in the tumor mass that inhibit the effector function of the anti-tumor specific T cells. Thus, the identification of immunomodulators that can increase the frequency and/or the infiltration of antitumor specific T cells while reducing the suppressive capacity of the tumor microenvironment is necessary to ensure the effectiveness of T cell immunotherapies. In this review, we discuss the potential of poly-ICLC as a multi-functional immune modulator for treating cancer and its impact on the 3 above mentioned barriers. We describe the unique capacity of poly-ICLC in stimulating 2 separate pattern recognition receptors, TLR3 and cytosolic MDA5 and the consequences of these activations on cytokines and chemokines production. We emphasize the role of poly-ICLC as an adjuvant in the setting of peptide-based cancer vaccines and in situ tumor vaccination by mimicking natural immune responses to infections. Finally, we summarize the impact of poly-ICLC in enhancing T infiltration into the tumor parenchyma and address the implication of this finding in the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Carboxymethylcellulose Sodium/analogs & derivatives , Immunologic Factors/pharmacology , Immunomodulation , Poly I-C/immunology , Poly I-C/pharmacology , Polylysine/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Carboxymethylcellulose Sodium/pharmacology , Carboxymethylcellulose Sodium/therapeutic use , Cytokines/metabolism , Humans , Immunity, Innate/drug effects , Immunologic Factors/therapeutic use , Immunomodulation/drug effects , Interferon-Induced Helicase, IFIH1/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Poly I-C/therapeutic use , Polylysine/immunology , Polylysine/pharmacology , Polylysine/therapeutic use , Receptors, Pattern Recognition/metabolism , Toll-Like Receptor 3/metabolism
3.
Dev Comp Immunol ; 106: 103635, 2020 05.
Article in English | MEDLINE | ID: mdl-32014470

ABSTRACT

Thioredoxin-like protein 5 (Trxlp-5) is a thioredoxin isoform associated with cellular redox homeostasis through the activity of thiol-disulfide reductase. In our study, Trxlp-5 was identified and characterized in Pinctada fucata martensii. The expression of PmTrxlp-5 was detected in response to polyinosinic: polycytidylic acid (poly I:C) and lipopolysaccharides (LPS) stimulation. The differences in PmTrxlp-5 expression were evaluated between the black coloured selected line and the control stock after grafting operation. The open reading frame (ORF) consisted of 1167bp encoding a 388 amino acid, 5'-UTR of 41bp and a 3'-UTR of 846bp. PmTrxlp-5 exhibited a conserved WCXXC functional motif similar to thioredoxins from other species. Tissue analysis showcased the highest relative mRNA expressions of PmTrxlp-5 in the haemocytes. Interestingly, after the grafting operation, mRNA expression of PmTrxlp-5 in the haemocytes was differentially expressed post grafting with a peak 6 h after grafting suggesting the high involvement of the gene in immune response in the early stage after grafting. The black coloured selected line group (BS) had significantly higher expression than the control group (CG) at 24 h, 6 d and 30 d after grafting operation. PmTrxlp-5 also showed a wave-like pattern in mRNA expression after bacterial endotoxin LPS and viral mimic poly I:C. These results suggested that PmTrxlp-5 plays a vital function in cellular redox homeostasis and immune response against grafting operation and pathogenic infections and can be used as a gene marker for selective breeding programs.


Subject(s)
Hemocytes/physiology , Pinctada/immunology , Thioredoxins/genetics , Transplantation, Homologous , Animals , Cloning, Molecular , Genetic Markers , Humans , Immunity, Innate/genetics , Lipopolysaccharides/immunology , Phylogeny , Poly I-C/immunology , Polylysine/immunology , Selective Breeding , Thioredoxins/metabolism , Transcriptome
4.
Urol Clin North Am ; 47(4S): e1-e8, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33446322

ABSTRACT

Relatively simple, synthetic, double-stranded RNAs can be powerful viral pathogen-associated molecular pattern (PAMP) mimics, inducing a panoply of antiviral and antitumor responses that act at multiple stages of host defense. Their mechanisms of action and uses are beginning to be understood, alone, in combination with other therapeutics, or as novel PAMP-adjuvants providing the critical danger signal that has been missing from most cancer and other modern vaccines. Dose, timing, route of administration combinations, and other clinical variables can have a critical impact on immunogenicity. This article reviews advances in the use of polyinosinic-polycytidylic acid and derivatives, in particular poly-ICLC.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Carboxymethylcellulose Sodium/analogs & derivatives , Immunologic Factors/therapeutic use , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Prostatic Neoplasms/therapy , RNA, Double-Stranded/therapeutic use , Adjuvants, Immunologic/physiology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Carboxymethylcellulose Sodium/therapeutic use , Clinical Trials as Topic , Humans , Immunologic Factors/immunology , Male , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/therapeutic use , Poly I-C/immunology , Polylysine/immunology , Polylysine/therapeutic use , Prostatic Neoplasms/immunology , RNA, Double-Stranded/immunology
5.
Front Immunol ; 11: 569331, 2020.
Article in English | MEDLINE | ID: mdl-33505391

ABSTRACT

The LabEx Milieu Interieur (MI) project is a clinical study centered on the detailed characterization of the baseline and induced immune responses in blood samples from 1,000 healthy donors. Analyses of these samples has lay ground for seminal studies on the genetic and environmental determinants of immunologic variance in a healthy cohort population. In the current study we developed in vitro methods enabling standardized quantification of MI-cohort-derived primary fibroblasts responses. Our results show that in vitro human donor cohort fibroblast responses to stimulation by different MAMPs analogs allows to characterize individual donor immune-phenotype variability. The results provide proof-of-concept foundation to a new experimental framework for such studies. A bio-bank of primary fibroblast lines was generated from 323 out of 1,000 healthy individuals selected from the MI-study cohort. To study inter-donor variability of innate immune response in primary human dermal fibroblasts we chose to measure the TLR3 and TLR4 response pathways, both receptors being expressed and previously studied in fibroblasts. We established high-throughput automation compatible methods for standardized primary fibroblast cell activation, using purified MAMPS analogs, poly I:C and LPS that stimulate TLR3 and TLR4 pathways respectively. These results were in turn compared with a stimulation method using infection by HSV-1 virus. Our "Add-only" protocol minimizes high-throughput automation system variability facilitating whole process automation from cell plating through stimulation to recovery of cell supernatants, and fluorescent labeling. Images were acquired automatically by high-throughput acquisition on an automated high-content imaging microscope. Under these methodological conditions standardized image acquisition provided for quantification of cellular responses allowing biological variability to be measured with low system noise and high biological signal fidelity. Optimal for automated analysis of immuno-phenotype of primary human cell responses our method and experimental framework as reported here is highly compatible to high-throughput screening protocols like those necessary for chemo-genomic screening. In context of primary fibroblasts derived from donors enrolled to the MI-clinical-study our results open the way to assert the utility of studying immune-phenotype characteristics relevant to a human clinical cohort.


Subject(s)
Biological Variation, Population/immunology , Fibroblasts/immunology , Fibroblasts/metabolism , Host-Pathogen Interactions/immunology , Immunity, Innate , Biological Assay/methods , Cell Line , Cells, Cultured , Cytokines/metabolism , Female , Flow Cytometry , Gene Expression , Genes, Reporter , Herpesvirus 1, Human/immunology , Humans , Lipopolysaccharides/immunology , Middle Aged , Poly I-C/immunology , Polylysine/immunology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
6.
Malar J ; 18(1): 146, 2019 Apr 24.
Article in English | MEDLINE | ID: mdl-31014347

ABSTRACT

BACKGROUND: Plasmodium falciparum parasite is the most deadly species of human malaria, and the development of an effective vaccine that prevents P. falciparum infection and transmission is a key target for malarial elimination and eradication programmes. P. falciparum cell-traversal protein for ookinetes and sporozoites (PfCelTOS) is an advanced vaccine candidate. A comparative study was performed to characterize the immune responses in BALB/c mouse immunized with Escherichia coli-expressed recombinant PfCelTOS (rPfCelTOS) in toll-like receptor (TLR)-based adjuvants, CpG and Poly I:C alone or in combination (CpG + Poly I:C), followed by the assessment of transmission-reducing activity (TRA) of anti-rPfCelTOS antibodies obtained from different vaccine groups in Anopheles stephensi. METHODS: The aim of the current work was achieved by head-to-head comparison of the vaccine groups using conventional and avidity enzyme-linked immunosorbent assay (ELISA), immunofluorescence test (IFAT), and standard membrane feeding assay (SMFA). RESULTS: Comparing to rPfCelTOS alone, administration of rPfCelTOS with two distinct TLR-based adjuvants in vaccine mouse groups showed a significant increase in responses (antibody level, IgG subclass analysis, avidity, and Th1 cytokines) and was able to induce reasonable transmission-reducing activity. Also, comparable functional activity of anti-rPfCelTOS antibodies was found in group that received antigen in either CpG or Poly I:C (69.9%/20% and 73.5%/24.4%, respectively, reductions in intensity/prevalence). However, the vaccine group receiving rPfCelTOS in combination with CpG + Poly I:C showed a significant induction in antibody titers and inhibitory antibodies in oocysts development (78.3%/19.6% reductions in intensity/prevalence) in An. stephensi. CONCLUSIONS: A key finding in this investigation is that rPfCelTOS administered alone in BALB/c mouse is poorly immunogenic, with relatively low IgG level, avidity, inhibitory antibodies, and mixed Th1/Th2 responses. However, immunological characteristic (IgG level, cytophilic IgG2a and IgG2b, avidity, and Th1 cytokines) and TRA of anti-rPfCelTOS significantly enhanced in the presence of co-administration of TLR-based adjuvants, confirming that targeting TLRs would be an effective means for the enhancement of inducing TRA against rPfCelTOS.


Subject(s)
Anopheles/parasitology , Antibodies, Protozoan/blood , Antibody Affinity , Malaria, Falciparum/prevention & control , Protozoan Proteins/immunology , Toll-Like Receptors/immunology , Adjuvants, Immunologic , Animals , Antigens, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Immunization , Immunoglobulin G/blood , Malaria Vaccines/immunology , Malaria, Falciparum/transmission , Mice, Inbred BALB C , Oligodeoxyribonucleotides/immunology , Oocysts , Plasmodium falciparum/immunology , Poly I-C/immunology , Polylysine/immunology , Sporozoites/immunology
7.
Front Immunol ; 9: 259, 2018.
Article in English | MEDLINE | ID: mdl-29515577

ABSTRACT

Devil facial tumor disease (DFTD) is renowned for its successful evasion of the host immune system. Down regulation of the major histocompatabilty complex class I molecule (MHC-I) on the DFTD cells is a primary mechanism of immune escape. Immunization trials on captive Tasmanian devils have previously demonstrated that an immune response against DFTD can be induced, and that immune-mediated tumor regression can occur. However, these trials were limited by their small sample sizes. Here, we describe the results of two DFTD immunization trials on cohorts of devils prior to their wild release as part of the Tasmanian Government's Wild Devil Recovery project. 95% of the devils developed anti-DFTD antibody responses. Given the relatively large sample sizes of the trials (N = 19 and N = 33), these responses are likely to reflect those of the general devil population. DFTD cells manipulated to express MHC-I were used as the antigenic basis of the immunizations in both trials. Although the adjuvant composition and number of immunizations differed between trials, similar anti-DFTD antibody levels were obtained. The first trial comprised DFTD cells and the adjuvant combination of ISCOMATRIX™, polyIC, and CpG with up to four immunizations given at monthly intervals. This compared to the second trial whereby two immunizations comprising DFTD cells and the adjuvant combination ISCOMATRIX™, polyICLC (Hiltonol®) and imiquimod were given a month apart, providing a shorter and, therefore, more practical protocol. Both trials incorporated a booster immunization given up to 5 months after the primary course. A key finding was that devils in the second trial responded more quickly and maintained their antibody levels for longer compared to devils in the first trial. The different adjuvant combination incorporating the RNAase resistant polyICLC and imiquimod used in the second trial is likely to be responsible. The seroconversion in the majority of devils in these anti-DFTD immunization trials was remarkable, especially as DFTD is hallmarked by its immune evasion mechanisms. Microsatellite analyzes of MHC revealed that some MHC-I microsatellites correlated to stronger immune responses. These trials signify the first step in the long-term objective of releasing devils with immunity to DFTD into the wild.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines/immunology , Facial Neoplasms/immunology , Immunotherapy/methods , Marsupialia/immunology , Animals , Carboxymethylcellulose Sodium/analogs & derivatives , Female , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Imiquimod/immunology , Immunity, Humoral , Immunization, Secondary , Immunoglobulin G/blood , Male , Poly I-C/immunology , Polylysine/analogs & derivatives , Polylysine/immunology , Tumor Escape
8.
Clin Cancer Res ; 24(5): 1019-1029, 2018 03 01.
Article in English | MEDLINE | ID: mdl-28947565

ABSTRACT

Purpose: Treatment options are limited for patients with high-risk myelodysplastic syndrome (MDS). The azanucleosides, azacitidine and decitabine, are first-line therapy for MDS that induce promoter demethylation and gene expression of the highly immunogenic tumor antigen NY-ESO-1. We demonstrated that patients with acute myeloid leukemia (AML) receiving decitabine exhibit induction of NY-ESO-1 expression in circulating blasts. We hypothesized that vaccinating against NY-ESO-1 in patients with MDS receiving decitabine would capitalize upon induced NY-ESO-1 expression in malignant myeloid cells to provoke an NY-ESO-1-specific MDS-directed cytotoxic T-cell immune response.Experimental Design: In a phase I study, 9 patients with MDS received an HLA-unrestricted NY-ESO-1 vaccine (CDX-1401 + poly-ICLC) in a nonoverlapping schedule every four weeks with standard-dose decitabine.Results: Analysis of samples serially obtained from the 7 patients who reached the end of the study demonstrated induction of NY-ESO-1 expression in 7 of 7 patients and NY-ESO-1-specific CD4+ and CD8+ T-lymphocyte responses in 6 of 7 and 4 of 7 of the vaccinated patients, respectively. Myeloid cells expressing NY-ESO-1, isolated from a patient at different time points during decitabine therapy, were capable of activating a cytotoxic response from autologous NY-ESO-1-specific T lymphocytes. Vaccine responses were associated with a detectable population of CD141Hi conventional dendritic cells, which are critical for the uptake of NY-ESO-1 vaccine and have a recognized role in antitumor immune responses.Conclusions: These data indicate that vaccination against induced NY-ESO-1 expression can produce an antigen-specific immune response in a relatively nonimmunogenic myeloid cancer and highlight the potential for induced antigen-directed immunotherapy in a group of patients with limited options. Clin Cancer Res; 24(5); 1019-29. ©2017 AACRSee related commentary by Fuchs, p. 991.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Cancer Vaccines/administration & dosage , Decitabine/administration & dosage , Immunotherapy/methods , Leukemia, Myeloid, Acute/therapy , Myelodysplastic Syndromes/therapy , Aged , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cancer Vaccines/immunology , Carboxymethylcellulose Sodium/administration & dosage , Carboxymethylcellulose Sodium/analogs & derivatives , Combined Modality Therapy/methods , Dose-Response Relationship, Drug , Drug Administration Schedule , Feasibility Studies , Humans , Interferon Inducers/administration & dosage , Interferon Inducers/immunology , Leukemia, Myeloid, Acute/immunology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Membrane Proteins/metabolism , Middle Aged , Myelodysplastic Syndromes/immunology , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Poly I-C/administration & dosage , Poly I-C/immunology , Polylysine/administration & dosage , Polylysine/analogs & derivatives , Polylysine/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Treatment Outcome
9.
J Immunol ; 200(2): 688-703, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29203516

ABSTRACT

In mammals, RIG-I like receptors (RLRs) RIG-I and melanoma differentiation-associated gene 5 (MDA5) sense cytosolic viral RNA, leading to IFN antiviral response; however, LGP2 exhibits controversial functions. The same happens to fish LGP2. In this study we report that three zebrafish LGP2 splicing transcripts, a full-length LGP2, and two truncating variants, LGP2v1 and LGP2v2, play distinct roles during IFN antiviral response. Overexpression of the full-length LGP2 not only potentiates IFN response through the RLR pathway, in the absence or presence of poly(I:C) at limited concentrations, but also inhibits IFN response by relative high concentrations of poly(I:C) through functional attenuation of signaling factors involved in the RLR pathway; however, LGP2v1 and LGP2v2 only retain the inhibitory role. Consistently, LGP2 but not LGP2v1 and LGP2v2 confers protection on fish cells against spring viremia of carp virus (SVCV) infection and at limited expression levels, LGP2 exerts more significant protection than either RIG-I or MDA5. Further data suggest that in the early phase of SVCV infection, LGP2 functions as a positive regulator but along with SVCV replicating in cells up to a certain titer, which leads to a far more robust expression of IFN, LGP2 switches to a negative role. These in vitro results suggest an ingenious mechanism where the three zebrafish LGP2 splicing transcripts work cooperatively to shape IFN antiviral responses.


Subject(s)
Alternative Splicing , Antiviral Agents/metabolism , Disease Resistance/genetics , Fish Proteins/genetics , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Interferons/biosynthesis , Zebrafish/genetics , Amino Acid Sequence , Animals , Cell Line , Disease Resistance/immunology , Fish Diseases/genetics , Fish Diseases/immunology , Fish Diseases/virology , Host-Pathogen Interactions/immunology , Poly I-C/immunology , Polylysine/immunology , RNA Helicases/genetics , RNA Helicases/metabolism , Signal Transduction , Virus Replication/immunology , Zebrafish/virology
10.
Vaccine ; 35(51): 7139-7146, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29153777

ABSTRACT

Human parainfluenza virus type 3 (PIV3) is a major cause of lower respiratory disease i.e. bronchitis, bronchiolitis or pneumonia, in infants and young children. Presently there is no licensed vaccine against PIV3. To produce an effective subunit vaccine, a chimeric FHN glycoprotein consisting of the N-terminal ectodomain of the fusion (F) protein linked to the haemagglutinin-neuraminidase (HN) protein without transmembrane domain, and secreted forms of the individual F and HN glycoproteins, were expressed in mammalian cells and purified. Mice and cotton rats were immunized intramuscularly (IM) with FHN or both F and HN proteins (F + HN), formulated with poly(I:C) and an innate defense regulator peptide in polyphosphazene (TriAdj). Significantly higher levels of systemic virus-neutralizing antibodies were observed in mice and cotton rats immunized with FHN/TriAdj when compared to animals immunized with the combination of F and HN proteins (F + HN/TriAdj). As PIV3 is a pneumotropic virus, another goal is to produce an effective mucosal subunit vaccine. Intranasal (IN) administration with FHN/TriAdj resulted in mucosal IgA production in the lung and virus neutralizing antibodies in the sera. After PIV3 challenge no virus was detected in cotton rats immunized with FHN/TriAdj regardless of the route of delivery. Protective immunity against PIV3 was also induced by FHN/TriAdj in hamsters. In conclusion, the FHN protein formulated with TriAdj has potential for development of a safe and effective vaccine against PIV3.


Subject(s)
Adjuvants, Immunologic/administration & dosage , HN Protein/immunology , Parainfluenza Virus 3, Human/immunology , Viral Fusion Proteins/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Cricetinae , HN Protein/administration & dosage , HN Protein/genetics , Humans , Immunization , Mice , Poly I-C/administration & dosage , Poly I-C/immunology , Polylysine/administration & dosage , Polylysine/immunology , Sigmodontinae , Vaccination , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/adverse effects , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Viral Fusion Proteins/administration & dosage , Viral Fusion Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/adverse effects
11.
Int J Mol Med ; 40(5): 1495-1503, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28949380

ABSTRACT

Toll-like receptor 3 (TLR3) and TLR4 utilize adaptor proteins to activate mitogen­activated protein kinase (MAPK), resulting in the acute but transient inflammatory response aimed at the clearance of pathogens. In the present study, it was demonstrated that macrophage activation by lipopolysaccharide (LPS) or poly(I:C), leading to changes in cell morphology, differed significantly between the mouse macrophage cell line RAW264.7 and mouse primary peritoneal macrophages. Moreover, the expression of α- and ß-tubulin was markedly decreased following LPS stimulation. By contrast, α- and ß-tubulin expression were only mildly increased following poly(I:C) treatment. However, the expression of ß-actin and GAPDH was not significantly affected. Furthermore, it was verified that vincristine pretreatment abrogated the cytoskeleton rearrangement and decreased the synthesis and secretion of proinflammatory cytokines and migration of macrophages caused by LPS. Finally, it was observed that the MAPK/p38 signaling pathway regulating cytoskeleton rearrangement may participate in LPS­induced macrophage cytokine production and migration. Overall, the findings of the present study indicated that MAPK/p38 regulation of the cytoskeleton, particularly tubulin proteins, plays an important role in LPS-induced inflammatory responses via alleviating the synthesis and secretion of proinflammatory cytokines and inhibiting the migration of macrophages.


Subject(s)
Cytoskeleton/metabolism , MAP Kinase Signaling System , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Cytokines/metabolism , Dose-Response Relationship, Drug , Lipopolysaccharides/immunology , Male , Mice , Poly I-C/immunology , Polylysine/immunology , RAW 264.7 Cells , Tubulin Modulators/pharmacology , Vincristine/pharmacology
12.
Dev Comp Immunol ; 76: 352-360, 2017 11.
Article in English | MEDLINE | ID: mdl-28689773

ABSTRACT

Devil facial tumour disease (DFTD) describes two genetically distinct transmissible tumours that pose a significant threat to the survival of the Tasmanian devil. A prophylactic vaccine could protect devils from DFTD transmission. For this vaccine to be effective, potent immune adjuvants will be required. Toll-like receptors (TLRs) promote robust immune responses in human cancer studies and are highly conserved across mammalian species. In this study, we investigated the proficiency of TLR ligands for immune activation in the Tasmanian devil using in vitro mononuclear cell stimulations and in vivo immunisation trials with a model antigen. We identified two such TLR ligands, polyICLC (Hiltonol®) (TLR3) and imiquimod (TLR7), that in combination induced significant IFNγ production from Tasmanian devil lymphocytes in vitro. Immunisation with these ligands and the model antigen keyhole limpet haemocyanin activated robust antigen-specific primary, secondary and long-term memory IgG responses. Our results support the conserved nature of TLR signaling across mammalian species. PolyICLC and imiquimod will be trialed as immune adjuvants in future DFTD vaccine formulations.


Subject(s)
Aminoquinolines/immunology , Antigens/immunology , Cancer Vaccines/immunology , Carboxymethylcellulose Sodium/analogs & derivatives , Facial Neoplasms/immunology , Leukocytes, Mononuclear/immunology , Marsupialia/immunology , Poly I-C/immunology , Polylysine/analogs & derivatives , Adjuvants, Immunologic , Animals , Cells, Cultured , Facial Neoplasms/prevention & control , Hemocyanins/immunology , Humans , Imiquimod , Immunity , Immunity, Innate , Immunization , Immunoglobulin G/metabolism , Lymphocyte Activation , Polylysine/immunology , Toll-Like Receptors/agonists
13.
J Virol ; 91(9)2017 05 01.
Article in English | MEDLINE | ID: mdl-28202751

ABSTRACT

We compared the HIV-1-specific immune responses generated by targeting HIV-1 envelope protein (Env gp140) to either CD40 or LOX-1, two endocytic receptors on dendritic cells (DCs), in rhesus macaques primed with a poxvirus vector (NYVAC-KC) expressing Env gp140. The DC-targeting vaccines, humanized recombinant monoclonal antibodies fused to Env gp140, were administered as a boost with poly-ICLC adjuvant either alone or coadministered with the NYVAC-KC vector. All the DC-targeting vaccine administrations with poly-ICLC increased the low-level serum anti-Env IgG responses elicited by NYVAC-KC priming significantly more (up to a P value of 0.01) than in a group without poly-ICLC. The responses were robust and cross-reactive and contained antibodies specific to multiple epitopes within gp140, including the C1, C2, V1, V2, and V3, C4, C5, and gp41 immunodominant regions. The DC-targeting vaccines also elicited modest serum Env-specific IgA responses. All groups gave serum neutralization activity limited to tier 1 viruses and antibody-dependent cytotoxicity responses (ADCC) after DC-targeting boosts. Furthermore, CD4+ and CD8+ T cell responses specific to multiple Env epitopes were strongly boosted by the DC-targeting vaccines plus poly-ICLC. Together, these results indicate that prime-boost immunization via NYVAC-KC and either anti-CD40.Env gp140/poly-ICLC or anti-LOX-1.Env gp140/poly-ICLC induced balanced antibody and T cell responses against HIV-1 Env. Coadministration of NYVAC-KC with the DC-targeting vaccines increased T cell responses but had minimal effects on antibody responses except for suppressing serum IgA responses. Overall, targeting Env to CD40 gave more robust T cell and serum antibody responses with broader epitope representation and greater durability than with LOX-1.IMPORTANCE An effective vaccine to prevent HIV-1 infection does not yet exist. An approach to elicit strong protective antibody development is to direct virus protein antigens specifically to dendritic cells, which are now known to be the key cell type for controlling immunity. In this study, we have tested in nonhuman primates two prototype vaccines engineered to direct the HIV-1 coat protein Env to dendritic cells. These vaccines bind to either CD40 or LOX-1, two dendritic cell surface receptors with different functions and tissue distributions. We tested the vaccines described above in combination with attenuated virus vectors that express Env. Both vaccines, but especially that delivered via CD40, raised robust immunity against HIV-1 as measured by monitoring potentially protective antibody and T cell responses in the blood. The safety and efficacy of the CD40-targeted vaccine justify further development for future human clinical trials.


Subject(s)
AIDS Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Antibodies/immunology , HIV-1/immunology , Scavenger Receptors, Class E/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/genetics , Animals , Antibodies, Neutralizing/immunology , CHO Cells , Carboxymethylcellulose Sodium/analogs & derivatives , Cricetulus , Dendritic Cells/immunology , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Macaca mulatta , Male , Poly I-C/immunology , Polylysine/analogs & derivatives , Polylysine/immunology , Vaccination
14.
Sci Rep ; 6: 35113, 2016 10 12.
Article in English | MEDLINE | ID: mdl-27731424

ABSTRACT

The optimal recognition of penicillin determinants, including amoxicillin (AX), by specific IgE antibodies is widely believed to require covalent binding to a carrier molecule. The nature of the carrier and its contribution to the antigenic determinant is not well known. Here we aimed to evaluate the specific-IgE recognition of different AX-derived structures. We studied patients with immediate hypersensitivity reactions to AX, classified as selective or cross-reactors to penicillins. Competitive immunoassays were performed using AX itself, amoxicilloic acid, AX bound to butylamine (AXO-BA) or to human serum albumin (AXO-HSA) in the fluid phase, as inhibitors, and amoxicilloyl-poli-L-lysine (AXO-PLL) in the solid-phase. Two distinct patterns of AX recognition by IgE were found: Group A showed a higher recognition of AX itself and AX-modified components of low molecular weights, whilst Group B showed similar recognition of both unconjugated and conjugated AX. Amoxicilloic acid was poorly recognized in both groups, which reinforces the need for AX conjugation to a carrier for optimal recognition. Remarkably, IgE recognition in Group A (selective responders to AX) is influenced by the mode of binding and/or the nature of the carrier; whereas IgE in Group B (cross-responders to penicillins) recognizes AX independently of the nature of the carrier.


Subject(s)
Amoxicillin/adverse effects , Amoxicillin/immunology , Drug Hypersensitivity/immunology , Hypersensitivity, Immediate/immunology , Immunoglobulin E/blood , Adult , Aged , Amoxicillin/analogs & derivatives , Anaphylaxis/blood , Anaphylaxis/etiology , Anaphylaxis/immunology , Antibody Specificity , Butylamines/immunology , Carrier Proteins/blood , Carrier Proteins/immunology , Cross Reactions , Drug Hypersensitivity/blood , Drug Hypersensitivity/etiology , Female , Haptens/adverse effects , Haptens/immunology , Humans , Hypersensitivity, Immediate/blood , Hypersensitivity, Immediate/etiology , Male , Middle Aged , Penicillins/adverse effects , Penicillins/immunology , Polylysine/immunology , Serum Albumin, Human/immunology , Urticaria/blood , Urticaria/etiology , Urticaria/immunology , Young Adult , beta-Lactams/adverse effects , beta-Lactams/immunology
15.
J Neurooncol ; 130(3): 517-527, 2016 12.
Article in English | MEDLINE | ID: mdl-27624914

ABSTRACT

Recurrent high-grade gliomas (HGGs) of childhood have an exceedingly poor prognosis with current therapies. Accordingly, new treatment approaches are needed. We initiated a pilot trial of vaccinations with peptide epitopes derived from glioma-associated antigens (GAAs) overexpressed in these tumors in HLA-A2+ children with recurrent HGG that had progressed after prior treatments. Peptide epitopes for three GAAs (EphA2, IL13Rα2, survivin), emulsified in Montanide-ISA-51, were administered subcutaneously adjacent to intramuscular injections of poly-ICLC every 3 weeks for 8 courses, followed by booster vaccines every 6 weeks. Primary endpoints were safety and T-cell responses against the GAA epitopes, assessed by enzyme-linked immunosorbent spot (ELISPOT) analysis. Treatment response was evaluated clinically and by magnetic resonance imaging. Twelve children were enrolled, 6 with glioblastoma, 5 with anaplastic astrocytoma, and one with malignant gliomatosis cerebri. No dose-limiting non-CNS toxicity was encountered. ELISPOT analysis, in ten children, showed GAA responses in 9: to IL13Rα2 in 4, EphA2 in 9, and survivin in 3. One child had presumed symptomatic pseudoprogression, discontinued vaccine therapy, and responded to subsequent treatment. One other child had a partial response that persisted throughout 2 years of vaccine therapy, and continues at >39 months. Median progression-free survival (PFS) from the start of vaccination was 4.1 months and median overall survival (OS) was 12.9 months. 6-month PFS and OS were 33 and 73 %, respectively. GAA peptide vaccination in children with recurrent malignant gliomas is generally well tolerated, and has preliminary evidence of immunological and modest clinical activity.


Subject(s)
Antigens, Neoplasm/immunology , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Glioma/therapy , Immunotherapy, Active/methods , Adolescent , Antigens, Neoplasm/chemistry , Carboxymethylcellulose Sodium/analogs & derivatives , Child , Child, Preschool , Female , Glioma/immunology , Glioma/metabolism , Humans , Infant , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/immunology , Interleukin-13 Receptor alpha1 Subunit , Male , Peptides/immunology , Pilot Projects , Poly I-C/immunology , Polylysine/analogs & derivatives , Polylysine/immunology , Receptor, EphA2/chemistry , Receptor, EphA2/immunology , Receptors, Interleukin-13/chemistry , Receptors, Interleukin-13/immunology , Survivin , Treatment Outcome , Young Adult
16.
Neuro Oncol ; 18(8): 1157-68, 2016 08.
Article in English | MEDLINE | ID: mdl-26984745

ABSTRACT

BACKGROUND: Low-grade gliomas (LGGs) are the most common brain tumors of childhood. Although surgical resection is curative for well-circumscribed superficial lesions, tumors that are infiltrative or arise from deep structures are therapeutically challenging, and new treatment approaches are needed. Having identified a panel of glioma-associated antigens (GAAs) overexpressed in these tumors, we initiated a pilot trial of vaccinations with peptides for GAA epitopes in human leukocyte antigen-A2+ children with recurrent LGG that had progressed after at least 2 prior regimens. METHODS: Peptide epitopes for 3 GAAs (EphA2, IL-13Rα2, and survivin) were emulsified in Montanide-ISA-51 and administered subcutaneously adjacent to intramuscular injections of polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose every 3 weeks for 8 courses, followed by booster vaccines every 6 weeks. Primary endpoints were safety and T-lymphocyte responses against GAA epitopes. Treatment response was evaluated clinically and by MRI. RESULTS: Fourteen children were enrolled. Other than grade 3 urticaria in one child, no regimen-limiting toxicity was encountered. Vaccination induced immunoreactivity to at least one vaccine-targeted GAA in all 12 evaluable patients: to IL-13Rα2 in 3, EphA2 in 11, and survivin in 3. One child with a metastatic LGG had asymptomatic pseudoprogression noted 6 weeks after starting vaccination, followed by dramatic disease regression with >75% shrinkage of primary tumor and regression of metastatic disease, persisting >57 months. Three other children had sustained partial responses, lasting >10, >31, and >45 months, and one had a transient response. CONCLUSIONS: GAA peptide vaccination in children with recurrent LGGs is generally well tolerated, with preliminary evidence of immunological and clinical activity.


Subject(s)
Antigens, Neoplasm/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Carboxymethylcellulose Sodium/analogs & derivatives , Glioma/drug therapy , Glioma/immunology , Interferon Inducers/therapeutic use , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Vaccination/methods , Adolescent , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/immunology , Carboxymethylcellulose Sodium/administration & dosage , Carboxymethylcellulose Sodium/adverse effects , Carboxymethylcellulose Sodium/therapeutic use , Child , Child, Preschool , Disease-Free Survival , Epitopes , Female , Humans , Infant , Inhibitor of Apoptosis Proteins/immunology , Interferon Inducers/administration & dosage , Interferon Inducers/adverse effects , Interferon Inducers/immunology , Interleukin-13 Receptor alpha2 Subunit/immunology , Male , Neoplasm Grading , Pilot Projects , Poly I-C/administration & dosage , Poly I-C/adverse effects , Poly I-C/immunology , Polylysine/administration & dosage , Polylysine/adverse effects , Polylysine/immunology , Polylysine/therapeutic use , Receptor, EphA2/immunology , Survivin , Treatment Outcome
17.
J Immunol Res ; 2016: 2643297, 2016.
Article in English | MEDLINE | ID: mdl-28127565

ABSTRACT

Background. T2 inflammation of chronic rhinosinusitis with nasal polyps (CRSwNP) may be influenced by epithelial cytokines release (TSLP, IL-25, and IL-33). We investigated the release of TSLP, IL-25, and IL-33 by epithelial CRSwNP cells compared to epithelial sinus mucosa cells of patients with chronic rhinosinusitis without nasal polyps (CRSsNP). Methods. IL-25, IL-33, and TSLP were measured by ELISA in the supernatant of cell cultures derived by CRSwNP (9 patients, 6 atopic) and CRSsNP (7 patients, 2 atopic) in baseline condition and following stimulation with Dermatophagoides pteronyssinus (DP), Aspergillus fumigatus (AF), and poly(I:C). Results. CRSwNP epithelial cells released increased levels of IL-25 (from 0.12 ± 0.06 pg/ml to 0.27 ± 0.1 pg/ml, p < 0.01) and TSLP (from 0.77 ± 0.5 pg/ml to 2.53 ± 1.17 pg/ml, p < 0.001) following poly(I:C) stimulation, while CRSsNP epithelial cells released increased levels of IL-25 and IL-33 following AF and DP stimulation, respectively (IL-25: from 0.18 ± 0.07 pg/ml to 0.51 ± 0.1 pg/ml, p < 0.001; IL-33: from 2.57 ± 1.3 pg/ml to 5.7 ± 3.1 pg/ml, p < 0.001). Conclusions. CRSwNP epithelial cells release TSLP and IL-25 when stimulated by poly(I:C) but not by DP or AF, suggesting that viral infection may contribute to maintain and amplify the T2 immune response seen in CRSwNP.


Subject(s)
Cytokines/metabolism , Interleukin-17/metabolism , Interleukin-33/metabolism , Nasal Mucosa/immunology , Nasal Polyps/immunology , Rhinitis/immunology , Sinusitis/immunology , Adult , Aged , Aged, 80 and over , Animals , Antigens, Dermatophagoides/immunology , Aspergillus fumigatus/immunology , Cells, Cultured , Chronic Disease , Culture Media , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/immunology , Female , Humans , Interleukin-17/immunology , Interleukin-33/immunology , Male , Middle Aged , Poly I-C/immunology , Polylysine/immunology , Young Adult , Thymic Stromal Lymphopoietin
18.
J Immunol ; 195(3): 1015-24, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26123354

ABSTRACT

Nonlive vaccine platforms that induce potent cellular immune responses in mucosal tissue would have broad application for vaccines against infectious diseases and tumors. Induction of cellular immunity could be optimized by targeted activation of multiple innate and costimulatory signaling pathways, such as CD40 or TLRs. In this study, we evaluated immune activation and elicitation of T cell responses in nonhuman primates after immunization with peptide Ags adjuvanted with an agonistic anti-CD40Ab, with or without the TLR3 ligand poly IC:LC. We found that i.v. administration of the anti-CD40Ab induced rapid and transient innate activation characterized by IL-12 production and upregulated costimulatory and lymph node homing molecules on dendritic cells. Using fluorescently labeled Abs for in vivo tracking, we found that the anti-CD40Ab bound to all leukocytes, except T cells, and disseminated to multiple organs. CD4(+) and CD8(+) T cell responses were significantly enhanced when the anti-CD40Ab was coadministered with poly IC:LC compared with either adjuvant given alone and were almost exclusively compartmentalized to the lung. Notably, Ag-specific T cells in the bronchoalveolar lavage were sustained at ∼5-10%. These data indicate that systemic administration of anti-CD40Ab may be particularly advantageous for vaccines and/or therapies that require T cell immunity in the lung.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Lung/immunology , Lymphocyte Activation , Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies/administration & dosage , Antibodies/immunology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Carboxymethylcellulose Sodium/administration & dosage , Carboxymethylcellulose Sodium/analogs & derivatives , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Immunity, Cellular/immunology , Interleukin-12/biosynthesis , Lung/cytology , Macaca mulatta , Poly I-C/administration & dosage , Poly I-C/immunology , Polylysine/administration & dosage , Polylysine/analogs & derivatives , Polylysine/immunology , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Vaccination
19.
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
20.
Clin Cancer Res ; 20(5): 1355-65, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24520093

ABSTRACT

PURPOSE: Myeloma-directed cellular immune responses after autologous stem cell transplantation (ASCT) may reduce relapse rates. We studied whether coinjecting the TLR-3 agonist and vaccine adjuvant Poly-ICLC with a MAGE-A3 peptide vaccine was safe and would elicit a high frequency of vaccine-directed immune responses when combined with vaccine-primed and costimulated autologous T cells. EXPERIMENTAL DESIGN: In a phase II clinical trial (NCT01245673), we evaluated the safety and activity of ex vivo expanded autologous T cells primed in vivo using a MAGE-A3 multipeptide vaccine (compound GL-0817) combined with Poly-ICLC (Hiltonol), granulocyte macrophage colony-stimulating factor (GM-CSF) ± montanide. Twenty-seven patients with active and/or high-risk myeloma received autografts followed by anti-CD3/anti-CD28-costimulated autologous T cells, accompanied by MAGE-A3 peptide immunizations before T-cell collection and five times after ASCT. Immune responses to the vaccine were evaluated by cytokine production (all patients), dextramer binding to CD8(+) T cells, and ELISA performed serially after transplant. RESULTS: T-cell infusions were well tolerated, whereas vaccine injection site reactions occurred in >90% of patients. Two of nine patients who received montanide developed sterile abscesses; however, this did not occur in the 18 patients who did not receive montanide. Dextramer staining demonstrated MAGE-A3-specific CD8 T cells in 7 of 8 evaluable HLA-A2(+) patients (88%), whereas vaccine-specific cytokine-producing T cells were generated in 19 of 25 patients (76%). Antibody responses developed in 7 of 9 patients (78%) who received montanide and only weakly in 2 of 18 patients (11%) who did not. The 2-year overall survival was 74% [95% confidence interval (CI), 54%-100%] and 2-year event-free survival was 56% (95% CI, 37%-85%). CONCLUSIONS: A high frequency of vaccine-specific T-cell responses were generated after transplant by combining costimulated autologous T cells with a Poly-ICLC/GM-CSF-primed MAGE-A3 vaccine.


Subject(s)
Antigens, Neoplasm/immunology , Carboxymethylcellulose Sodium/analogs & derivatives , Immunotherapy, Adoptive , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Neoplasm Proteins/immunology , Poly I-C/immunology , Polylysine/analogs & derivatives , T-Lymphocytes/immunology , Adult , Aged , Cancer Vaccines/immunology , Female , Hematopoietic Stem Cell Transplantation , Humans , Immunization , Immunotherapy, Adoptive/adverse effects , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/mortality , Polylysine/immunology , Transplantation, Autologous , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...