Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nanotechnology ; 25(35): 355101, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25119543

ABSTRACT

Mannan (MN) is the natural ligand for mannose receptors, which are widely expressed on dendritic cells (DCs). The purpose of this study was to assess the effect of formulation parameters on the immunogenicity of MN-decorated poly (D, L-lactide-co-glycolide) (PLGA) nanoparticles (NPs) in terms of their ability to stimulate DC phenotypic as well as functional maturation. For this purpose, NPs were formulated from either ester-terminated or COOH-terminated PLGA. Incorporation of MN in NPs was achieved through encapsulation, physical adsorption or chemical conjugation. Murine bone marrow derived DCs (BMDCs) were treated with various NP formulations and assessed for their ability to up-regulate DC cell surface markers, secrete immunostimulatory cytokines and to activate allogenic T cell responses. DCs treated with COOH-terminated PLGA-NPs containing chemically conjugated MN (MN-Cov-COOH) have shown superior performance in improving DC biological functions, compared to the rest of the formulations tested. This may be attributed to the higher level of MN incorporation in the former formulation. Incorporation of MN in PLGA NPs through chemical conjugation can lead to enhanced DC maturation and stimulatory function. This strategy may be used to develop more effective PLGA-based vaccine formulations.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunity/drug effects , Mannans/metabolism , Nanoparticles/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Biomarkers/metabolism , Chemistry, Pharmaceutical/methods , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunity/immunology , Lactic Acid/administration & dosage , Lactic Acid/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles/chemistry , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Up-Regulation/drug effects , Up-Regulation/immunology
2.
Transl Oncol ; 4(3): 178-88, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21633673

ABSTRACT

Persistent activation of STAT3 plays a major role in cancer progression and immune escape. Therefore, targeting STAT3 in tumors is essential to enhance/reactivate antitumor immune response. In our previous studies, we demonstrated the efficacy of stearic acid-modified polyethylenimine (PEI-StA) in promoting small interfering RNA (siRNA) silencing of STAT3 in B16.F10 melanoma in vitro and in vivo. In the current study, we examine the immunologic impact of this intervention. Toward this goal, the infiltration and activation of lymphocytes and dendritic cells (DCs) in the tumor mass were assessed using flow cytometry. Moreover, the levels of IFN-γ, IL-12, and TNF-α in homogenized tumor supernatants were determined. Moreover, mixed lymphocytes reaction using splenocytes of tumor-bearing mice was used to assess DC functionality on siRNA/lipopolyplexes intervention. Our results demonstrated up to an approximately fivefold induction in the infiltration of CD3(+) cells in tumor mass on STAT3 knockdown with high levels of CD4(+), CD8(+), and NKT cells. Consistently, DC infiltration in tumor milieu increased up to approximately fourfold. Those DCs were activated, in an otherwise suppressive microenvironment, as evidenced by a high expression of costimulatory molecules CD86 and CD40. ELISA analysis revealed a significant increase in IFN-γ, IL-12, and TNF-α. Moreover, mixed lymphocytes reaction demonstrated alloreactivity of these DCs as assessed by high T-cell proliferation and IL-2 production. Our results suggest a bystander immune response after local STAT3 silencing by siRNA. This strategy could be beneficial as an adjuvant therapy along with current cancer vaccine formulations.

3.
Adv Drug Deliv Rev ; 63(10-11): 943-55, 2011 Sep 10.
Article in English | MEDLINE | ID: mdl-21679733

ABSTRACT

Development of safe and effective cancer vaccine formulation is a primary focus in the field of cancer immunotherapy. The recognition of the crucial role of dendritic cells (DCs) in initiating anti-tumor immunity has led to the development of several strategies that target vaccine antigens to DCs as an attempt for developing potent, specific and lasting anti-tumor T cell responses. The main objective of this review is to provide an overview on the application of poly (d,l-lactic-co-glycolic acid) nanoparticles (PLGA-NPs) as cancer vaccine delivery system and highlight their potential in the development of future therapeutic cancer vaccines. PLGA-NPs containing antigens along with immunostimulatory molecules (adjuvants) can not only target antigen actively to DCs, but also provide immune activation and rescue impaired DCs from tumor-induced immuosupression.


Subject(s)
Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Drug Delivery Systems , Adjuvants, Immunologic/administration & dosage , Animals , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Dendritic Cells/metabolism , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Lactic Acid/chemistry , Nanoparticles , Neoplasms/immunology , Neoplasms/therapy , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , T-Lymphocytes/immunology
4.
Pharm Res ; 28(9): 2288-301, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21560020

ABSTRACT

PURPOSE: Mannosylation of vaccines is a promising strategy to selectively target vaccine antigens to the mannose receptor expressed on dendritic cells (DCs). The purpose of this study was to investigate the effect of mannan (MN) chemically conjugated to poly(D, L-lactide-co-glycolic acid) (PLGA) nanoparticles (NPs) on antigen-specific T-cell responses elicited by a model antigen (ovalbumin, OVA) loaded in PLGA-NPs. METHODS: In vitro T-cell proliferation assay was done to assess the ability of DCs treated with OVA-NPs (±MN decoration) to induce antigen-specific T-cell activation. The efficacy of this vaccination strategy was further evaluated in vivo, where T-cell proliferation was performed to evaluate activation of T-cell responses in lymph nodes and spleens isolated from the vaccinated mice. RESULTS: Our results demonstrate that MN-decorated antigen-loaded PLGA-NPs simultaneously enhanced antigen-specific CD4+ and CD8+ T-cell responses compared to non-decorated NPs. CONCLUSIONS: MN decoration of PLGA-NPs is a promising strategy for enhancing antigen-specific T-cell responses.


Subject(s)
Antigens/administration & dosage , Drug Carriers/chemistry , Lactic Acid/chemistry , Mannans/chemistry , Polyglycolic Acid/chemistry , T-Lymphocytes/immunology , Vaccines/administration & dosage , Animals , Antigens/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Dendritic Cells/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class II/genetics , Lectins, C-Type/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mannose Receptor , Mannose-Binding Lectins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Polylactic Acid-Polyglycolic Acid Copolymer , Receptors, Cell Surface/immunology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes/cytology , Vaccines/immunology
5.
Curr Drug Deliv ; 8(3): 261-73, 2011 May.
Article in English | MEDLINE | ID: mdl-21453265

ABSTRACT

Dendritic cells (DCs) are the key antigen presenting cells that link innate and adaptive immunity. In the periphery, DCs capture antigens, process them and migrate into the regional lymph nodes where they could initiate antigen specific T cell immune responses. Immunotherapeutic strategies that aim to deliver tumor antigens specifically to DCs could not only boost anti-tumor immune responses but also could alleviate non-specific immune activation and/or unwanted side effects. Nano-sized particulate delivery systems are efficient modalities that can deliver tumor antigens to DCs in a targeted and specific manner. This review will provide general information on the rationale behind targeting antigens to DCs and the crucial role of DCs in initiating antigen specific T cell responses. Different strategies that have been employed in delivering antigens to DCs will be also discussed. A special emphasis will be put on specific targeting of cancer vaccine formulations to DC-specific receptors (e.g. CD11c, CD40, Fcγ, CCR6, pathogenic recognition receptors such as Toll-like receptors (TLRs) and C-type lectin receptors (CLRs)).


Subject(s)
Immunotherapy/methods , Nanoparticles/administration & dosage , Neoplasms/immunology , Neoplasms/therapy , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Dendritic Cells/immunology , Humans
6.
AAPS J ; 13(2): 159-68, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21336534

ABSTRACT

We have previously developed micelles of methoxy poly(ethylene oxide)-b-poly(ε-caprolactone) as vehicles for the solubilization and delivery of cyclosporine A (CsA). These micelles were able to reduce the renal uptake and nephrotoxicity of CsA. The purpose of the current study was to test the efficacy of polymeric micellar formulation of CsA (PM-CsA) in suppressing immune responses by either T cells or dendritic cells (DCs). The performance of PM-CsA was compared to that of the commercially available formulation of CsA (Sandimmune®). Our results demonstrate that PM-CsA could exert a potent immunosuppressive effect similar to that of Sandimmune® both in vitro and in vivo. Both formulations inhibited phenotypic maturation of DCs and impaired their allostimulatory capacity. Furthermore, both PM-CsA and Sandimmune® have shown similar dose-dependent inhibition of in vitro T cell proliferative responses. A similar pattern was observed in the in vivo study, where T cells isolated from both PM-CsA-treated and Sandimmune®-treated mice have shown impairment in their proliferative response and IFN-γ production at similar levels. These results highlight the potential of polymeric micelles to serve as efficient vehicles for the delivery of CsA.


Subject(s)
Cyclosporine/pharmacology , Drug Carriers/chemistry , Immunosuppressive Agents/pharmacology , Polyesters/chemistry , Animals , Cell Proliferation/drug effects , Cyclosporine/administration & dosage , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dose-Response Relationship, Drug , Immunosuppressive Agents/administration & dosage , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Micelles , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
7.
Curr Drug Deliv ; 8(3): 274-81, 2011 May.
Article in English | MEDLINE | ID: mdl-21291375

ABSTRACT

The interaction of dendritic cells (DCs) and T cells has been the cornerstone of approaches to cancer immunotherapy. Antitumoral immune responses can be elicited by delivering cancer antigens to DCs. As antigen presenting cells, these DCs activate cancer antigen specific T cells. Whereas the first part of the review discusses methods for delivery of cancer vaccines to DCs, in this part the focus is on the potential role of nanoscopic devices for molecular imaging of these immune responses. Nanoscopic devices could potentially deliver tracking molecules to DCs, enabling monitoring of DCs and/or T cell activation and tumoricidal activity during immunotherapy, using non-invasive imaging modalities such as nuclear imaging (single photon emission computed tomography (SPECT), positron emission tomography (PET)), magnetic resonance imaging (MRI) and optical imaging.


Subject(s)
Immunotherapy/methods , Molecular Imaging/methods , Nanoparticles/therapeutic use , Neoplasms/immunology , Neoplasms/therapy , Animals , Dendritic Cells/immunology , Humans , T-Lymphocytes/immunology
8.
J Drug Target ; 19(4): 281-92, 2011 May.
Article in English | MEDLINE | ID: mdl-20590403

ABSTRACT

The purpose of this study was to identify an optimum targeted particulate formulation based on mannan (MN)-decorated poly(D, L-lactide-co-glycolide) (PLGA) nanoparticles (NPs), for efficient delivery of incorporated cargo to dendritic cells (DCs). In brief, NPs were formulated from two different types of PLGA; ester-terminated (capped) or COOH-terminated (uncapped) polymer. Incorporation of MN in NPs was achieved either through addition of MN during the process of NP formation or by attachment of MN onto the surface of the freeze dried NPs by physical adsorption or chemical conjugation (to COOH terminated polymer). The formulated NPs were characterized in terms of particle size, Zeta potential and level of MN incorporation. The effect of polymer type and the incorporation method on the extent of fluorescently labelled NP uptake by murine bone marrow-derived DCs have been investigated using flowcytometry. The results of this study showed MN incorporation to enhance the uptake of PLGA NPs by DCs. Among different MN incorporation strategies, covalent attachment of MN to COOH-terminated PLGA-NPs provided the highest level of MN surface decoration on NPs. Maximum NP uptake by DCs was achieved by COOH terminated PLGA NPs containing covalent or adsorbed MN. Therefore, a better chance of success for these formulations for active targeted drug and/or vaccine delivery to DCs is anticipated.


Subject(s)
Dendritic Cells/metabolism , Lactic Acid/chemistry , Lactic Acid/metabolism , Macrophages/metabolism , Mannans/chemistry , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Polyglycolic Acid/metabolism , Animals , Drug Delivery Systems , Mice , Mice, Inbred C57BL , Molecular Structure , Polylactic Acid-Polyglycolic Acid Copolymer
9.
Mol Pharm ; 7(5): 1643-54, 2010 Oct 04.
Article in English | MEDLINE | ID: mdl-20804176

ABSTRACT

In dendritic cells (DCs), the induction of signal transducer and activator of transcription 3 (STAT3) by tumor-derived factors (TDFs) renders DCs tolerogenic and suppresses their antitumor activity. Therefore, silencing STAT3 in DCs is beneficial for cancer immunotherapy. We have shown that STAT3 knockdown in B16 murine melanoma by siRNA polyplexes of polyethylenimine (PEI) or its stearic acid derivative (PEI-StA) induces B16 cell death in vitro and in vivo. Here, we investigated the physical encapsulation of siRNA/PEI and PEI-StA polyplexes in poly(d,l-lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) for STAT3 knockdown in DCs. PLGA NPs containing siRNA polyplexes of PEI (PLGA-P) and PEI-StA (PLGA-PS) had an average diameter of ~350 to 390 nm and a zeta potential of ∼-13 to -19 mV, respectively. The encapsulation efficiency (E.E.) of siRNA in PLGA-P and PLGA-PS was 26% and 43%, respectively. In both NP types, siRNA release followed a triphasic pattern, but it was faster in PLGA-PS. Our uptake study by fluorescence microscopy confirmed DC uptake and endosomal localization of both NP types. After exposure to B16.F10 conditioned medium, DCs showed high STAT3 and low CD86 expression indicating impaired function. STAT3 silencing by PLGA-P and PLGA-PS of STAT3 siRNA restored DC maturation and functionality as evidenced by the upregulation of CD86 expression, high secretion of TNF-α and significant allogenic T cell proliferation. Moreover, encapsulation in PLGA NPs significantly reduced PEI-associated toxicity on DCs. We propose this formulation as a strategy for targeted siRNA delivery to DCs. The potential of this approach is not limited to STAT3 downregulation in DCs but can be used to target the expression of other proteins as well. Moreover, it can be combined with other means for cancer immunotherapy like cancer vaccine strategies.


Subject(s)
Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Base Sequence , Biological Transport, Active , Cell Line, Tumor , Cells, Cultured , Dendritic Cells/immunology , Gene Knockdown Techniques , Lactic Acid , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Microscopy, Electron, Scanning , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Nanocapsules/ultrastructure , Nanotechnology , Polyethyleneimine , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology
10.
J Pharm Pharm Sci ; 13(1): 21-6, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20456827

ABSTRACT

PURPOSE: Tumors can escape immune eradication by harnessing dendritic cell (DC) maturation. However, DC types used as in vitro models to study tumor-mediated immunosuppression possess fundamental variability that could influence research outcomes. Therefore, we assessed the behavior of two distinct murine DC models upon exposure to tumor-conditioned medium of B16.F10 melanoma (B16-CM). METHODS: Using primary bone-marrow derived dendritic cells (BMDCs) or immortalized DC2.4 cell line, we evaluated the level of signal transducer and activator of transcription 3 (STAT3) phosphorylation by Western blot as a molecular parameter. We also examined the surface expression of co-stimulatory molecules on DCs by flow cytometry as a phenotypic parameter. RESULTS: Our results revealed critical discrepancies between the two models in response to tumor-conditioned medium. While conditioned medium was able to induce STAT3 phosphorylation in BMDCs, it did not significantly induce STAT3 phosphorylation in DC2.4 cell line. Moreover, only in BMDCs, the expression of CD86 and CD40 was remarkably downregulated by B16-CM and was not totally recovered after LPS stimulation. In contrast, DC2.4 cells did not show any signs of harnessed maturation upon exposure to B16-CM. CONCLUSIONS: In order to study the effect of tumor-mediated immunosuppression on DC maturation in vitro via tumor-induction of STAT3 activation, primary BMDCs are more reliable as a model than DC2.4.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance , Melanoma, Experimental/immunology , STAT3 Transcription Factor/immunology , Animals , B7-2 Antigen/genetics , Blotting, Western , Bone Marrow Cells/metabolism , CD40 Antigens/genetics , Cell Line , Down-Regulation , Flow Cytometry , Mice , Mice, Inbred C57BL , Models, Biological , Phosphorylation
11.
Biomaterials ; 31(6): 1420-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19913908

ABSTRACT

Persistent activation of signal transducer and activator of transcription 3 (STAT3) has been shown to impart several oncogenic features in many solid and blood tumors. In this study, we investigated the potential of nanoparticles based on polyethylenimine (PEI) modified with stearic acid (StA), to deliver siRNA for efficient STAT3 downregulation in B16 melanoma cells. The B16 cells were targeted with approximately 6-200 nm of siRNA complexes for 36 h. Compared to the PEI complexes, the PEI-StA complexes showed higher potency in STAT3 silencing in B16 cells accompanied by a significant induction of IL-6 secretion and a reduction of VEGF production. Moreover, with PEI-StA complexes, the level of the cellular Caspase 3 activity (an indicator of apoptotic activity) was found to be 2.5 times higher than that of PEI complexes. When the cells were treated with 50 nm of siRNA complexes on a daily basis, the cell viability was dramatically reduced reaching only to 16% after the third daily dose of PEI-StA complexes, as compared to the 69% viability observed with the PEI complexes at an equivalent time period. Consistently, in vivo results indicated significant regression in tumor growth and tumor weight after siRNA/PEI-StA treatment as compared to the siRNA/PEI. This was accompanied with significant increase in IL-6 levels and Caspase 3 activity, and a significant decrease in VEGF level and STAT3 activity in the tumor tissue. The lipid-modified PEI is a promising carrier for siRNA delivery and downregulation of STAT3 by polymer-mediated siRNA delivery is an effective strategy for cancer treatment especially when an optimum delivery system can potentiate the silencing activity of siRNA.


Subject(s)
Apoptosis/drug effects , Lipids/chemistry , Melanoma/genetics , Melanoma/therapy , Polyethyleneimine/chemistry , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , STAT3 Transcription Factor/genetics , Cell Line, Tumor , Drug Carriers/chemistry , Gene Silencing , Genetic Therapy/methods , Humans , Materials Testing
12.
Mol Pharm ; 7(2): 364-74, 2010 Apr 05.
Article in English | MEDLINE | ID: mdl-20030320

ABSTRACT

Constitutively activated signal transducer and activator of transcription-3 (STAT3) in tumor and dendritic cells (DCs) plays a critical role in tumor-induced immunosuppression. This is considered a major challenge in effective immunotherapy of cancer. Herein we describe the development of a polymeric nanocarrier for the delivery of JSI-124 (a small molecule inhibitor of STAT3) to tumor and immunosuppressed DCs using poly(d,l-lactic-co-glycolic acid) nanoparticles (PLGA NPs). For this purpose, JSI-124 was chemically conjugated to PLGA and the PLGA-JSI-124 conjugate was formulated into nanoparticles using the emulsification solvent evaporation method. The attachment of JSI-124 to PLGA was confirmed by a combination of thin layer chromatography and (1)H NMR. The level of JSI-124 in NPs, determined by liquid chromatography-mass spectrometry, was found to be 1.7 +/- 0.3 microg per mg of PLGA. The PLGA-JSI-124 NPs demonstrated a controlled drug release profile over a 1-month period and exhibited potent anticancer and STAT3 inhibitory activity comparable to the soluble JSI-124 after 24 h incubation with B16 melanoma cells, in vitro. Moreover, PLGA-JSI-124 NPs efficiently suppressed the level of p-STAT3 in p-STAT3(high) DCs, generated from mouse bone marrow cells in the presence of conditioned media of B16 cells (B16CM-DCs), and improved their function as assessed by mixed lymphocyte reaction (MLR). Specifically cotreatment of B16CM-DCs with PLGA-JSI-124 NPs and PLGA NPs containing the DC adjuvant CpG resulted in higher levels of T cell proliferation in the MLR assay compared with B16CM-DCs untreated or treated with either CpG NPs or JSI-124 NPs alone. Our results indicate that PLGA NPs containing conjugated JSI-124 can potentially provide a useful platform for sustained JSI-124 release in tumor and its targeted delivery to DCs leading to the modulation of anticancer response by JSI-124 in tumor cells and immunosuppressed DCs, in vitro.


Subject(s)
Immunotherapy/methods , Lactic Acid/chemistry , Nanoparticles/chemistry , Neoplasms/therapy , Polyglycolic Acid/chemistry , STAT3 Transcription Factor/antagonists & inhibitors , Triterpenes/chemistry , Triterpenes/therapeutic use , Animals , Cell Line, Tumor , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Flow Cytometry , Lymphocyte Culture Test, Mixed , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Models, Biological , Neoplasms/immunology , Polylactic Acid-Polyglycolic Acid Copolymer
13.
Immunopharmacol Immunotoxicol ; 31(2): 214-21, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18798092

ABSTRACT

The efficiency of cancer immunotherapy strategies is hampered by the existence of an intra-tumoral immunosuppressive environment involving tolerogenic dendritic cells (DCs) and regulatory T (T(reg)) cells. Hyperactivation of STAT3 in tumor is implicated in the generation of this immunosuppressive environment. The purpose of this study was to test whether simultaneous inhibition of STAT3 in tumor and TLR4 ligand-induced activation of DCs can modulate tumor-induced immunosuppression. For this purpose, the effects of a TLR4 ligand, 7-acyl lipid A, delivered by poly(lactic-co-glycolic acid) nanoparticles (PLGA-NPs) to DCs on the activity of DCs and T(reg) cells was evaluated in vitro. In addition the immunomodulatory and anticancer effects of 7-acyl lipid A PLGA-NPs in combination with a STAT3 inhibitory agent, JSI-124, in a B16 mouse melanoma model was explored, in vivo. PLGA-NP delivery of 7-acyl lipid A to DCs reduced the suppressive effects of T(reg) cells on T cells in vitro. Besides, daily Intra-tumoral co-administration of 7-acyl lipid A PLGA-NPs and JSI-124 in C57BL/6 mice bearing B16-F10 tumor for 8 days resulted in a significant increase in the percentage of tumor infiltrated T cells as compared with control group that received PBS and monotherapy groups. The average tumor volume in the tumor-bearing mice that received JSI-124 plus 7-acyl lipid A PLGA-NPs combination therapy was found to be significantly lower than that in PBS and monotherapy groups. Our findings show a potential for the combination of STAT3 inhibition in tumor and TLR4 induced DC activation in increasing the efficacy of cancer immunotherapy.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Delivery Systems , Immune Tolerance/drug effects , Lipid A/analogs & derivatives , Melanoma, Experimental/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Triterpenes/administration & dosage , Animals , Dendritic Cells/drug effects , Dendritic Cells/immunology , Glycolates/administration & dosage , Immune Tolerance/immunology , Injections, Intralesional , Lactic Acid , Lipid A/administration & dosage , Male , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
14.
Vaccine ; 26(39): 5046-57, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18680779

ABSTRACT

The purpose of this study was to evaluate the efficacy of poly(lactic-co-glycolic acid) (PLGA)-based vaccines in breaking immunotolerance to cancer-associated self-antigens. Vaccination of mice bearing melanoma B16 tumors with PLGA nanoparticles (NP) co-encapsulating the poorly immunogenic melanoma antigen, tyrosinase-related protein 2 (TRP2), along with Toll-like receptor (TLR) ligand (7-acyl lipid A) was examined. Remarkably, this vaccine was able to induce therapeutic anti-tumor effect. Activated TRP2-specific CD8 T cells were capable of interferon (IFN)-gamma secretion at lymph nodes and spleens of the vaccinated mice. More importantly, TRP2/7-acyl lipid A-NP treated group has shown immunostimulatory milieu at the tumor microenvironment, as evidenced by increased level of pro-inflammatory cytokines compared to control group. These results support the potential use of PLGA nanoparticles as competent carriers for future cancer vaccine formulations.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Lactic Acid/therapeutic use , Melanoma, Experimental/therapy , Polyglycolic Acid/therapeutic use , Toll-Like Receptor 4/immunology , Animals , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Drug Carriers/therapeutic use , Interferon-gamma/immunology , Intramolecular Oxidoreductases/immunology , Lactic Acid/administration & dosage , Lipid A/immunology , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Polyglycolic Acid/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer
15.
Immunol Cell Biol ; 86(6): 506-14, 2008.
Article in English | MEDLINE | ID: mdl-18392040

ABSTRACT

One of the major limitations for cancer immunotherapy is related to the frequent existence of an intra-tumoral immunosuppressive environment, to which STAT3 (Signal transducer and activator of transcription-3) activation in tumor and dendritic cells (DCs) are believed to contribute. In this study, we tested the hypothesis that the combination of CpG (a DC activator) and JSI-124 (a STAT3 inhibitor) may generate synergistic antitumor effects compared to CpG or JSI-124 alone. B16-F10, a mouse melanoma cell line that has constitutively active STAT3, was grafted in C57BL/6 mice and then tumor-bearing mice treated intra-tumorally with (a) phosphate buffered saline, (b) 10 microg CpG, (c) 1 mg kg(-1) JSI-124 or (d) 10 microg CpG+1 mg kg(-1) JSI-124. The effects of treatments on tumor growth, survival and antitumor immune responses were evaluated. Although significant antitumor effects were detected with the single-agent treatments, the CpG+JSI-124 treatment resulted in synergistic antitumor effects compared to CpG or JSI-124 alone. Correlating with these findings, the combination therapy resulted in significantly higher intra-tumoral levels of several proinflammatory, TH1-related cytokines (including IL-12, IFN-gamma, TNF-alpha and IL-2), increases in intra-tumoral CD8+ and CD4+ T cells expressing activation/memory markers and NK cells and increases in activated DCs in the tumors and regional lymph nodes (LNs). Concomitantly, the combination therapy led to a significantly decreased level of immunosuppression, as evidenced by lower intra-tumoral level of VEGF and TGF-beta, and decreased number of CD4+CD25+Foxp3+ regulatory T cells in the regional LNs. This study has provided the proof-of-principle for combining CpG and JSI-124 to enhance antitumor immune responses.


Subject(s)
Drug Synergism , Melanoma, Experimental/therapy , Oligodeoxyribonucleotides/therapeutic use , STAT3 Transcription Factor/antagonists & inhibitors , Triterpenes/therapeutic use , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Combined Modality Therapy , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interferon-gamma/metabolism , Interleukin-12/metabolism , Interleukin-2/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Tumor Necrosis Factor-alpha/metabolism
16.
J Pharm Biomed Anal ; 44(4): 914-23, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17590559

ABSTRACT

The present study had two main objectives. First, was to compare the immune stimulatory effect of two synthetic lipid A analogues (7-acyl lipid A and pentaerythritol-based lipid A (PET lipid A)) on maturation/stimulation of bone marrow derived dendritic cells (DCs). Our second objective was to develop a liquid chromatography/mass spectrometry (LC-MS) method for the quantitative analysis of lipid A-based vaccine adjuvants. Treatment of immature DCs with 7-acyl lipid A and PET lipid A up regulated the surface expression of CD86 and CD40 molecules, and also induced similar profile of pro-inflammatory cytokine secretion. LC-MS analyses were performed using a Waters Micromass ZQ 4000 spectrometer, coupled to a Waters 2795 separations module with an autosampler. Calibration curves with R(2)>0.999 were constructed over the concentration range of 1.25-20 microg/ml for the solution of 7-acyl lipid A and PET lipid A. The method was tested in a 3 day validation protocol. The accuracy of the assay at different concentrations tested ranged from 89 to 108% and from 92 to 107% for 7-acyl lipid A and PET lipid A, respectively. The limit of quantification for both 7-acyl lipid A and PET lipid A was 1.25 microg/ml (signal/noise (S/N)) ratio >15:1. The sensitivity of the method (the limit of detection) was 0.35 and 0.15 ng for 7-acyl lipid A and PET lipid A, respectively (S/N ratio between 4:1 or 3:1). As a preliminary application, this method has been successfully applied to the determination of 7-acyl lipid A and PET lipid A content in poly (D,L-lactic-co-glycolic acid) nanoparticles (PLGA-NP).


Subject(s)
Adjuvants, Immunologic/analysis , Lactic Acid/analysis , Lipid A/analysis , Nanoparticles/analysis , Polyglycolic Acid/analysis , Polymers/analysis , Animals , B7-2 Antigen/biosynthesis , Bone Marrow Cells/physiology , CD40 Antigens/biosynthesis , Chemistry, Pharmaceutical , Chromatography, Liquid , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dendritic Cells/physiology , Drug Compounding , Flow Cytometry , Lipid A/analogs & derivatives , Mass Spectrometry , Mice , Mice, Inbred C57BL , Phenotype , Polylactic Acid-Polyglycolic Acid Copolymer , Propylene Glycols/chemistry , Reference Standards , Reproducibility of Results , Solutions , Up-Regulation/drug effects
17.
J Immunother ; 30(4): 378-95, 2007.
Article in English | MEDLINE | ID: mdl-17457213

ABSTRACT

A clinically relevant delivery system that can efficiently target and deliver antigens and adjuvant to dendritic cells (DCs) is under active investigation. Immunization with antigens and immunomodulators encapsulated in poly(D,L-lactic-co-glycolic acid) (PLGA) nanoparticles elicits potent cellular immune responses; but understanding how this mode of delivery affects DCs and priming of naive T cells needs further investigation. In the current study, we assessed the extent of maturation of DCs after treatment with monophosphoryl lipid A (MPLA) encapsulated in PLGA nanoparticles and the generation of primary T-cell immune responses elicited by DCs loaded with antigens using this approach. Results indicated that DCs up-regulated the expression of surface maturation markers and demonstrated an enhanced allostimulatory capacity after treatment with MPLA containing PLGA nanoparticles. Treatment of DCs with MPLA containing nanoparticles released high amounts of proinflammatory and TH1 (T helper 1) polarizing cytokines and chemokines greater than that achieved by MPLA in solution. The delivery of ovalbumin in PLGA nanoparticles to DCs induced potent in vitro and in vivo antigen-specific primary TH1 immune responses that were furthermore enhanced with codelivery of MPLA along with the antigen in the nanoparticle formulation. Delivery of MUC1 lipopeptide (BLP25, a cancer vaccine candidate) and MPLA in PLGA nanoparticles to human DCs induced proliferation of MUC1 reactive T cells in vitro demonstrating the break in tolerance to self-antigen MUC1. These results demonstrated that targeting antigens along with toll-like receptor ligands in PLGA nanoparticles to DCs is a promising approach for generating potent TH1 polarizing immune responses that can potentially override self-tolerance mechanisms and become beneficial in the immunotherapy of cancer and infectious diseases.


Subject(s)
Cancer Vaccines/chemistry , Dendritic Cells/immunology , Drug Delivery Systems , Immunotherapy/methods , Nanoparticles/chemistry , Animals , Cell Proliferation , Dendritic Cells/metabolism , Lactic Acid/chemistry , Lipid A/analogs & derivatives , Lipid A/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers/chemistry , Th1 Cells/immunology , Th2 Cells/immunology , Up-Regulation
18.
J Biomed Mater Res A ; 81(3): 652-62, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17187395

ABSTRACT

The purpose of this research was to investigate the use of biodegradable poly(D,L-lactic-co-glycolic acid) nanoparticles (PLGA-NP) as a vaccine delivery system to codeliver antigen, ovalbumin (OVA) along with monophosphoryl lipid A (MPLA) as adjuvant for induction of potent CD4(+) and CD8(+) T cell responses. The primary CD4(+) T responses to OVA/MPLA NP were investigated using OVA-specific T cells from DO11.10 transgenic mice. Following adoptive transfer of these cells, mice were immunized s.c. by NP formulations. For assessing the CD8(+) responses, bone marrow derived dendritic cells (DCs) were pulsed with different OVA formulations, then, cocultured with CD8(+) T cells from OT-1 mice. T cell proliferation/activation and IFN-gamma secretion profile have been examined. Particulate delivery of OVA and MPLA to the DCs lead to markedly increase in in vitro CD8(+) T cell T cell proliferative responses (stimulation index >3000) and >13-folds increase in in vivo clonal expanded CD4(+) T cells. The expanded T cells were capable of cytokine secretion and expressed an activation and memory surface phenotype (CD62L(lo), CD11a(hi), and CD44(hi)). Codelivery of antigen and MPLA in PLGA-NP offers an effective method for induction of potent antigen specific CD4(+) and CD8(+) T cell responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Glycolates/administration & dosage , Lipid A/analogs & derivatives , Nanoparticles/administration & dosage , Ovalbumin/administration & dosage , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Clone Cells , Cytokines/metabolism , Epitopes/drug effects , Glycolates/pharmacology , Injections, Intraperitoneal , Injections, Subcutaneous , Kinetics , Lactic Acid , Lipid A/administration & dosage , Lipid A/pharmacology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/pharmacology , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Solubility/drug effects , Toll-Like Receptors/immunology , Vaccination
19.
J Pharm Pharm Sci ; 9(2): 158-64, 2006.
Article in English | MEDLINE | ID: mdl-16959185

ABSTRACT

PURPOSE: To develop a liquid chromatography/mass spectrometry (LC-MS) method for the quantitative analysis of cucurbitacin I (JSI-124), an anti-cancer inhibitor of the janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway, in rat plasma samples. METHODS: Standard samples of cucurbitacin I were prepared from a stock solution (1 mg/mL) in methanol. Internal standard (I.S.) was 4-hydroxybenzophenone. Extraction of cucurbitacin I and I.S. from rat plasma was performed using acetonitrile/dichloromethane. LC-MS analyses were performed using a Waters Micromass ZQ 4000 spectrometer, and chromatographic separation was achieved using a Waters XTerraMSC18 3.5 microm (2.1 x 50 mm) column as the stationary phase. The mobile phase consisting of a mixture of acetonitrile: water containing 1% formic acid with initial ratio of 20:80, employing a linear gradient to a final ratio of 40:60 v/v over 13 minutes, was delivered at a constant flow rate of 0.2 mL/min. The mass spectrometer was operated in negative ionization mode and analytes were quantified with single ion recording (SIR) at m/z 559 for cucurbitacin I and m/z 196.8 for I.S. RESULTS: Calibration curves with r2 > 0.999 were constructed over the concentration range of 5-10000 ng/mL for the solution of cucurbitacin I in methanol and 10-1000 ng/mL for rat plasma samples. The extraction recoveries were 86 and 98% for 50 ng/mL and 1000 ng/mL plasma concentration of cucurbitacin I, respectively. The intra- and inter-day coefficients of variation were less than 15%, and mean intraday errors were less than 10% at plasma concentration extending from 10-1000 ng/mL. CONCLUSION: The developed assay is sensitive, specific, reproducible and reliable for quantitative analysis of cucurbitacin I. Application in a pharmacokinetic assessment was proven in the rats given the drug.


Subject(s)
Antineoplastic Agents/blood , Chromatography, Liquid/methods , Mass Spectrometry/methods , Plasma/chemistry , Triterpenes/blood , Animals , Rats , Rats, Sprague-Dawley , Reproducibility of Results , STAT3 Transcription Factor/metabolism , Sensitivity and Specificity
20.
Br J Clin Pharmacol ; 55(6): 560-9, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12814450

ABSTRACT

AIMS: The goal of this study was to determine the frequencies of important allelic variants in the TPMT, NAT2, GST, SULT1A1 and MDR-1 genes in the Egyptian population and compare them with the frequencies in other ethnic populations. METHODS: Genotyping was carried out in a total of 200 unrelated Egyptian subjects. TPMT*2 was detected using an allele-specific polymerase chain reaction (PCR) assay. TPMT*3C and NAT2 variants (*5,*6 and *7) were detected using an allele-specific real-time PCR assay. Detection of GSTM1 and GSTT1 null alleles was performed simultaneously using a multiplex PCR assay. Finally, a PCR-restriction fragment length polymorphism assay was applied for the determination of TPMT*3A (*3B), SULT1A1*2 and MDR-1 (3435T) variants. RESULTS: Genotyping of TPMT revealed frequencies of 0.003 and 0.013 for TPMT*3A and TPMT*3C, respectively. No TPMT*2 or *3B was detected in the analysed samples. The frequencies of specific NAT2 alleles were 0.215, 0.497, 0.260 and 0.028 for *4 (wild-type), *5 (341C), *6 (590A) and *7 (857A), respectively. GSTM1 and GSTT1 null alleles were detected in 55.5% and 29.5% of the subjects, respectively. SULT1A1*2 was detected at a frequency of 0.135. Finally, the frequencies of the wild-type allele (3435C) and the 3435T variant in the MDR-1 gene were found to be 0.6 and 0.4, respectively. CONCLUSIONS: We found that Egyptians resemble other Caucasians with regard to allelic frequencies of the tested variants of NAT2, GST and MDR-1. By contrast, this Egyptian population more closely resemble Africans with respect to the TPMT*3C allele, and shows a distinctly different frequency with regard to the SULT1A1*2 variant. The predominance of the slow acetylator genotype in the present study (60.50%) could not confirm a previously reported higher frequency of the slow acetylator phenotype in Egyptians (92.00%), indicating the possibility of the presence of other mutations not detectable as T341C, G590A and G857A. The purpose of our future studies is to investigate for new polymorphisms, which could be relatively unique to the Egyptian population.


Subject(s)
Arylsulfotransferase , Genes, MDR/genetics , Arylamine N-Acetyltransferase/genetics , Base Pair Mismatch , Base Sequence , Egypt , Ethnicity , Gene Frequency , Genotype , Glutathione Transferase/genetics , Humans , Methyltransferases/genetics , Molecular Sequence Data , Sulfotransferases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...