Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
J Immunother Cancer ; 9(6)2021 06.
Article in English | MEDLINE | ID: mdl-34088742

ABSTRACT

BACKGROUND: NY-ESO-1 is a tumor-specific, highly immunogenic, human germ cell antigen of the MAGE-1 family that is a promising vaccine and cell therapy candidate in clinical trial development. The mouse genome does not encode an NY-ESO-1 homolog thereby not subjecting transgenic T-cells to thymic tolerance mechanisms that might impair in-vivo studies. We hypothesized that an NY-ESO-1 T cell receptor (TCR) transgenic mouse would provide the unique opportunity to study avidity of TCR response against NY-ESO-1 for tumor vaccine and cellular therapy development against this clinically relevant and physiological human antigen. METHODS: To study in vitro and in vivo the requirements for shaping an effective T cell response against the clinically relevant NY-ESO-1, we generated a C57BL/6 HLA-A*0201 background TCR transgenic mouse encoding the 1G4 TCR specific for the human HLA-A2 restricted, NY-ESO-1157-165 SLLMWITQC (9C), initially identified in an NY-ESO-1 positive melanoma patient. RESULTS: The HLA-A*0201 restricted TCR was positively selected on both CD4+ and CD8+ cells. Mouse 1G4 T cells were not activated by endogenous autoimmune targets or a large library of non-cognate viral antigens. In contrast, their activation by HLA-A2 NY-ESO-1157-165 complexes was evident by proliferation, CD69 upregulation, interferon-γ production, and interleukin-2 production, and could be tuned using a twofold higher affinity altered peptide ligand, NY-ESO-1157-165V. NY-ESO-1157-165V recombinant vaccination of syngeneic mice adoptively transferred with m1G4 CD8+ T cells controlled tumor growth in vivo. 1G4 transgenic mice suppressed growth of syngeneic methylcholanthrene (MCA) induced HHD tumor cells expressing the full-length human NY-ESO-1 protein but not MCA HHD tumor cells lacking NY-ESO-1. CONCLUSIONS: The 1G4 TCR mouse model for the physiological human TCR against the clinically relevant antigen, NY-ESO-1, is a valuable tool with the potential to accelerate clinical development of NY-ESO-1-targeted T-cell and vaccine therapies.


Subject(s)
HLA-A2 Antigen/metabolism , Neoplasm Proteins/administration & dosage , Peptide Fragments/administration & dosage , Receptors, Antigen, T-Cell/genetics , Thymoma/drug therapy , Thymus Neoplasms/drug therapy , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/immunology , Thymoma/genetics , Thymoma/immunology , Thymus Neoplasms/genetics , Thymus Neoplasms/immunology , Xenograft Model Antitumor Assays
2.
Oncogene ; 40(2): 334-344, 2021 01.
Article in English | MEDLINE | ID: mdl-33128043

ABSTRACT

We previously showed that Livin, an inhibitor of apoptosis protein, is specifically cleaved to produce a truncated protein, tLivin, and demonstrated its paradoxical proapoptotic activity. We further demonstrated that mini-tLivin (MTV), a 70 amino acids derivative of tLivin, is a proapoptotic protein as potent as tLivin. Based on these findings, in this study we aimed to develop a venue to target MTV for the treatment of diffuse large B-cell lymphoma (DLBCL). MTV was conjugated to poly (lactide-co-glycolic acid) surface-activated nanoparticles (NPs). In order to target MTV-NPs we also conjugated CD40 ligand (CD40L) to the surface of the NPs and evaluated the efficacy of the bifunctional CD40L-MTV-NPs. In vitro, CD40L-MTV-NPs elicited significant apoptosis of DLBCL cells. In a disseminated mouse model of DLBCL, 37.5% of MTV-NPs treated mice survived at the end of the experiment. Targeting MTV-NPs using CD40L greatly improved survival and 71.4% of these mice survived. CD40L-MTV-NPs also greatly reduced CNS involvement of DLBCL. Only 20% of these mice presented infiltration of lymphoma to the brain in comparison to 77% of the MTV-NPs treated mice. In a subcutaneous mouse model, CD40L-MTV-NPs significantly reduced tumor volume in correlation with significant increased caspase-3 activity. Thus, targeted MTV-NPs suggest a novel approach to overcome apoptosis resistance in cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/administration & dosage , Gene Expression Regulation, Neoplastic , Inhibitor of Apoptosis Proteins/administration & dosage , Lymphoma, Large B-Cell, Diffuse/therapy , Nanoparticles/administration & dosage , Neoplasm Proteins/administration & dosage , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis , Cell Proliferation , Humans , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Nanoparticles/chemistry , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Tumor Burden , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Eur Urol Focus ; 6(2): 280-283, 2020 03 15.
Article in English | MEDLINE | ID: mdl-30219710

ABSTRACT

Towards the development of vaccines against urinary tract infections (UTI), we determined the ability of intramuscular (i.m.) immunization to result in antigen-specific antibodies in urine. As a model antigen/vaccine, levels of total and vaccine-specific antibodies were determined in urine as a spin-out study of a phase 1 trial. Non-muscle-invasive bladder cancer (NMIBC) patients at different risks of progression, undergoing intravesical bacillus Calmette-Guérin (BCG) immunotherapy or not, received an adjuvanted recombinant protein vaccine that resulted in high titers of vaccine-specific serum immunoglobulin G (IgG) in all patients, regardless of the risk group. Vaccine-specific IgG and immunoglobulin A (IgA) were detected in urine of half of the patients at low risk of progression NMIBC and in all the intermediary/high- (int/high) risk patients. Vaccine-specific IgG titers were correlated to total urinary IgG levels, the latter being higher in the int/high-risk patients. In contrast, vaccine-specific IgA did not correlate to urinary IgA levels. Furthermore, vaccine-specific antibodies were transiently increased by intravesical BCG instillations. Altogether, our data show that a standard i.m. immunization can effectively induce antigen-specific antibodies in urine, which, upon selection of optimal vaccine targets, may provide protection against UTI. Vaccine-specific IgG titers were dependent on conditions affecting total urinary IgG levels, while production of vaccine-specific IgA in situ might independently contribute to protection against infections in the bladder. PATIENT SUMMARY: Towards the development of vaccines able to protect against urinary tract infections, we examined the potential of the intramuscular vaccination using a model antigen. We found two types of specific antibodies in the urine, which together may locally contribute to protection against infections, thus supporting the use of such a standard immunization route.


Subject(s)
Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , Immunization/methods , Immunoglobulin A/urine , Immunoglobulin G/urine , Neoplasm Proteins/administration & dosage , Neoplasm Proteins/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Adjuvants, Immunologic/therapeutic use , BCG Vaccine/therapeutic use , Humans , Injections, Intramuscular , Urinary Bladder Neoplasms/drug therapy , Urinary Tract Infections/prevention & control
6.
BMC Cancer ; 18(1): 1274, 2018 Dec 19.
Article in English | MEDLINE | ID: mdl-30567529

ABSTRACT

BACKGROUND: HDIL-2 is approved for advanced melanoma based on its durable antitumor activity. MAGE-A3 cancer immunotherapeutic (MAGE-A3 CI) is a recombinant MAGE-A3 protein combined with an immunostimulant adjuvant system and has shown antitumor activity in melanoma. We assessed the safety and anti-tumor activity of HDIL-2 combined with MAGE-A3 CI in advanced melanoma. METHODS: Patients with unresectable Stage III or Stage IV MAGE-A3-positive melanoma were enrolled in this phase II study. Treatment included an induction phase of MAGE-A3 CI plus HDIL-2 for 8 cycles followed by a maintenance phase of MAGE-A3 CI monotherapy. The primary endpoints were safety and objective response assessed per RECIST v1.1. Immune biomarker and correlative studies on tumor and peripheral blood were performed. RESULTS: Eighteen patients were enrolled. Seventeen patients were evaluable for safety and sixteen for response. Responses occurred in 4/16 (25%) patients with 3 complete responses, and stable disease in 6/16 (38%) patients with a disease control rate of 63%. The median duration of response was not reached at median follow-up of 36.8 months. Induction therapy of HDIL-2 + MAGE-A3 CI had similar toxicities to those reported with HDIL-2 alone. Maintenance MAGE-A3 monotherapy was well-tolerated. Increased immune checkpoint receptor expression by circulating T regulatory cells was associated with poor clinical outcomes; and responders tended to have increased tumor infiltrating T cells in the baseline tumor samples. CONCLUSIONS: The safety profile of HDIL-2 + MAGE-A3 CI was similar to HDIL-2 monotherapy. Maintenance MAGE-A3 CI provides robust anti-tumor activity in patients who achieved disease control with induction therapy. Immune monitoring data suggest that MAGE-A3 CI plus checkpoint inhibitors could be a promising treatment for MAGE-A3-positive melanoma. TRIAL REGISTRATION: ClinicalTrials.gov, NCT01266603 . Registered 12/24/2010, https://clinicaltrials.gov/ct2/show/NCT01266603.


Subject(s)
Antigens, Neoplasm/administration & dosage , Interleukin-2/administration & dosage , Melanoma/drug therapy , Neoplasm Proteins/administration & dosage , Recombinant Proteins/administration & dosage , Adjuvants, Immunologic , Adult , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Female , Humans , Immunotherapy , Interleukin-2/genetics , Interleukin-2/immunology , Male , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Staging , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Treatment Outcome
7.
Sci Adv ; 4(10): eaat8131, 2018 10.
Article in English | MEDLINE | ID: mdl-30402539

ABSTRACT

Intracellular delivery of mRNA, DNA, and other large macromolecules into cells plays an essential role in an array of biological research and clinical therapies. However, current methods yield a wide variation in the amount of material delivered, as well as limitations on the cell types and cargoes possible. Here, we demonstrate quantitatively controlled delivery into a range of primary cells and cell lines with a tight dosage distribution using a nanostraw-electroporation system (NES). In NES, cells are cultured onto track-etched membranes with protruding nanostraws that connect to the fluidic environment beneath the membrane. The tight cell-nanostraw interface focuses applied electric fields to the cell membrane, enabling low-voltage and nondamaging local poration of the cell membrane. Concurrently, the field electrophoretically injects biomolecular cargoes through the nanostraws and into the cell at the same location. We show that the amount of material delivered is precisely controlled by the applied voltage, delivery duration, and reagent concentration. NES is highly effective even for primary cell types or different cell densities, is largely cargo agnostic, and can simultaneously deliver specific ratios of different molecules. Using a simple cell culture well format, the NES delivers into >100,000 cells within 20 s with >95% cell viability, enabling facile, dosage-controlled intracellular delivery for a wide variety of biological applications.


Subject(s)
Cell Membrane/metabolism , Drug Delivery Systems , Green Fluorescent Proteins/administration & dosage , Nanostructures/administration & dosage , Nanotechnology/methods , Neoplasm Proteins/administration & dosage , RNA, Messenger/administration & dosage , Stromal Interaction Molecule 1/administration & dosage , Electroporation , HEK293 Cells , Humans , Nanostructures/chemistry
8.
IUBMB Life ; 70(10): 1002-1011, 2018 10.
Article in English | MEDLINE | ID: mdl-30171788

ABSTRACT

Therapeutic human papillomaviruse (HPV) vaccines have the potential to inhibit the tumor growth by targeting HPV E6 and E7 oncoproteins. Among different vaccine strategies, DNA and protein-based approaches are the most effective candidates for stimulation of the immune responses against HPV infections. Our study was designed to assess the efficacy of small heat shock proteins B1 (Hsp27) and B6 (Hsp20) as an adjuvant accompanied by HPV16 E7 and hPP10-E7 antigens in tumor mouse model. A major key for successful DNA and protein transfer into cells is the development of delivery systems with high efficiency and low cytotoxicity. Herein, we used hPP10 and MPG cell penetrating peptides (CPPs) for protein and DNA delivery in vivo, respectively. Our data indicated that the combination of Hsp27 with the recombinant hPP10-E7 protein in homologous protein/protein (hPP10-E7 + Hsp27) and heterologous DNA/protein (pcDNA-E7 + MPG/ hPP10-E7 + Hsp27) significantly enhanced the E7-specific T cell responses. Indeed, these regimens induced high levels of IgG2a, IFN-γ and IL-2 directed toward Th1 responses and also Granzyme B secretion as compared to other immunization strategies, and also displayed complete protection more than 60 days after treatment. These data suggest that the use of Hsp27 as an adjuvant and MPG and hPP10 as a gene and protein carrier would represent promising applications for improvement of HPV therapeutic vaccines. © 2018 IUBMB Life, 70(10):1002-1011, 2018.


Subject(s)
HSP20 Heat-Shock Proteins/genetics , Heat-Shock Proteins/administration & dosage , Neoplasm Proteins/administration & dosage , Papillomavirus Vaccines/administration & dosage , Uterine Cervical Neoplasms/genetics , Adjuvants, Immunologic/administration & dosage , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Cell Line, Tumor , Cell-Penetrating Peptides/administration & dosage , DNA-Binding Proteins/administration & dosage , Female , Granzymes/administration & dosage , Heat-Shock Proteins/genetics , Humans , Molecular Chaperones , Neoplasm Proteins/genetics , Papillomavirus E7 Proteins/administration & dosage , Papillomavirus Vaccines/genetics , Papillomavirus Vaccines/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology
10.
PLoS One ; 13(5): e0198039, 2018.
Article in English | MEDLINE | ID: mdl-29795667

ABSTRACT

Loss of integrity of the blood-brain barrier (BBB) in ischemic stroke victims initiates a devastating cascade of events causing brain damage. Maintaining the BBB is important to preserve brain function in ischemic stroke. Unfortunately, recombinant tissue plasminogen activator (tPA), the only effective fibrinolytic treatment at the acute stage of ischemic stroke, also injures the BBB and increases the risk of brain edema and secondary hemorrhagic transformation. Thus, it is important to identify compounds that maintain BBB integrity in the face of ischemic injury in patients with stroke. We previously demonstrated that intravenously injected phosphorylated recombinant heat shock protein 27 (prHSP27) protects the brains of mice with transient middle cerebral artery occlusion (tMCAO), an animal stroke-model. Here, we determined whether prHSP27, in addition to attenuating brain injury, also decreases BBB damage in hyperglycemic tMCAO mice that had received tPA. After induction of hyperglycemia and tMCAO, we examined 4 treatment groups: 1) bovine serum albumin (BSA), 2) prHSP27, 3) tPA, 4) tPA plus prHSP27. We examined the effects of prHSP27 by comparing the BSA and prHSP27 groups and the tPA and tPA plus prHSP27 groups. Twenty-four hours after injection, prHSP27 reduced infarct volume, brain swelling, neurological deficits, the loss of microvessel proteins and endothelial cell walls, and mortality. It also reduced the rates of hemorrhagic transformation, extravasation of endogenous IgG, and MMP-9 activity, signs of BBB damage. Therefore, prHSP27 injection attenuated brain damage and preserved the BBB in tPA-injected, hyperglycemic tMCAO experimental stroke-model mice, in which the BBB is even more severely damaged than in simple tMCAO mice. The attenuation of brain damage and BBB disruption in the presence of tPA suggests the effectiveness of prHSP27 and tPA as a combination therapy. prHSP27 may be a novel therapeutic agent for ischemic stroke patients whose BBBs are injured following tPA injections.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Ischemia/drug therapy , Heat-Shock Proteins/administration & dosage , Infarction, Middle Cerebral Artery/drug therapy , Neoplasm Proteins/administration & dosage , Neuroprotective Agents/administration & dosage , Stroke/drug therapy , Tissue Plasminogen Activator/adverse effects , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Ischemia/pathology , Heat-Shock Proteins/metabolism , Infarction, Middle Cerebral Artery/pathology , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Molecular Chaperones , Neoplasm Proteins/metabolism , Neuroprotective Agents/metabolism , Phosphorylation , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Stroke/pathology
11.
J Biol Chem ; 293(27): 10561-10573, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29802199

ABSTRACT

Host recognition of intracellular viral RNA and subsequent induction of cytokine signaling are tightly regulated at the cellular level and are a target for manipulation by viruses and therapeutics alike. Here, we characterize chromosome 6 ORF 106 (C6orf106) as an evolutionarily conserved inhibitor of the innate antiviral response. C6orf106 suppresses the synthesis of interferon (IFN)-α/ß and proinflammatory tumor necrosis factor (TNF) α in response to the dsRNA mimic poly(I:C) and to Sendai virus infection. Unlike canonical inhibitors of antiviral signaling, C6orf106 blocks interferon-regulatory factor 3 (IRF3) and, to a lesser extent, NF-κB activity without modulating their activation, nuclear translocation, cellular expression, or degradation. Instead, C6orf106 interacts with IRF3 and inhibits IRF3 recruitment to type I IFN promoter sequences while also reducing the nuclear levels of the coactivator proteins p300 and CREB-binding protein (CBP). In summary, we have defined C6orf106 as a negative regulator of antiviral immunity that blocks IRF3-dependent cytokine production via a noncanonical and poorly defined mechanism. This work presents intriguing implications for antiviral immunity, autoimmune disorders, and cancer.


Subject(s)
Antiviral Agents/pharmacology , Immunity, Innate/immunology , Interferon Regulatory Factor-3/antagonists & inhibitors , Neoplasm Proteins/pharmacology , Respirovirus Infections/prevention & control , Sendai virus/immunology , Animals , Antiviral Agents/administration & dosage , Chlorocebus aethiops , Gene Expression Regulation , HeLa Cells , Humans , Immunity, Innate/drug effects , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/administration & dosage , Respirovirus Infections/immunology , Respirovirus Infections/virology , Sendai virus/drug effects , Signal Transduction , Vero Cells
12.
Clin Exp Pharmacol Physiol ; 45(2): 140-145, 2018 02.
Article in English | MEDLINE | ID: mdl-28945941

ABSTRACT

Multiple myeloma (MM) is one of the most common hematological malignancies and characterized by the clonal accumulation of malignant plasma cells. Significant progress has been made in MM treatment recently, while MM still remains incurable. Our previous studies showed that the recombined human programmed cell death 5 (rhPDCD5) can promote MM apoptosis induced by dexamethasone (Dex). Here, we expanded the findings by showing that the rhPDCD5 alone could not induce an obvious growth inhibition of U266 cells (a MM cell line). Of note, with the combination of dexamethasone (Dex), the growth of MM cells was significantly inhibited and accompanied with the cell cycle arrest in G0/G1. For mechanism study, we found that the combination treatment of rhPDCD5 plus Dex downregulated the mRNA and protein expressions of Wnt effectors including ß-catenin, ß-catenin (Ser675), TCF4, survivin and c-Myc when compared to Dex only. Moreover, the activation of WNT pathway induced by LiCl can also be inhibited by this combination treatment. Taken together, our study demonstrated that the combination of rhPDCD5 and Dex can suppress the proliferation of multiple myeloma cells partially via inhibiting the WNT signalling pathway.


Subject(s)
Apoptosis Regulatory Proteins/pharmacology , Dexamethasone/pharmacology , Multiple Myeloma/drug therapy , Neoplasm Proteins/pharmacology , Wnt Proteins/metabolism , Apoptosis Regulatory Proteins/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Dexamethasone/administration & dosage , Drug Therapy, Combination , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lithium Chloride/pharmacology , Neoplasm Proteins/administration & dosage , Recombinant Proteins , Wnt Proteins/genetics
13.
Biomed Pharmacother ; 92: 108-115, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28531799

ABSTRACT

AIM: The aim of this study was to explain the Numb anti-cancer effects in the prostatic cancer. METHODS: Collecting the 20 prostatic cancer patients and analyzing the correlation between Numb and Glease score. Transfection Numb into DU-145 and PC-3 cells, measuring the proliferation rate of difference groups by MTT assay, evaluating the cell apoptosis and cell cycle of difference group by Flow cytometry; measuring the invasion and migration abilities by transwell and wound healing assays. In the nude mice experiment, establish prostatic cancer nude mouse subcutaneous planting tumor model by DU-145 cells, Injection the Numb from tail vein. Evaluating the tumor volume and weight. RESULTS: The Numb protein expression was decreased with Glease score increasing. The proliferation rate of Numb groups were significantly decreased compared with NC groups (P<0.05, respectively). The apoptosis and G1 phase rates of Numb groups were significantly enhanced compared with NC groups (P<0.05, respectively). The invasion and migration abilities of Numb group cells were significantly weaken compared with NC groups (P<0.05, respectively). In the WB assay, The relative proteins (Numb, P53, Cyclin D1, Rac1, MMP-2 and MMP-9) expression were significantly differences between NC and Numb groups (P<0.05, respectively). In the vivo experiment, the tumor volume and weight of Numb group was significantly lighter than NC group (P<0.05, respectively). CONCLUSION: Overexpression Numb had anti-cancer effects to prostatic cancer in vitro and vivo experiments, the mechanism might be P53/Cyclin D1 and Rac1/MMP-2/-9 signaling pathway.


Subject(s)
Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Prostatic Neoplasms/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , G1 Phase/drug effects , Humans , Injections, Intravenous , Male , Membrane Proteins/administration & dosage , Membrane Proteins/genetics , Membrane Proteins/therapeutic use , Mice, Nude , Neoplasm Grading , Neoplasm Invasiveness/prevention & control , Neoplasm Proteins/administration & dosage , Neoplasm Proteins/genetics , Neoplasm Proteins/therapeutic use , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
Vaccine ; 35(11): 1509-1516, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28196735

ABSTRACT

GTL001 is a bivalent therapeutic vaccine containing human papillomavirus (HPV) 16 and HPV18 E7 proteins inserted in the Bordetella pertussis adenylate cyclase (CyaA) vector intended to prevent cervical cancer in HPV-infected women with normal cervical cytology or mild abnormalities. To be effective, therapeutic cervical cancer vaccines should induce both a T cell-mediated effector response against HPV-infected cells and a robust CD8+ T-cell memory response to prevent potential later infection. We examined the ability of GTL001 and related bivalent CyaA-based vaccines to induce, in parallel, effector and memory CD8+ T-cell responses to both vaccine antigens. Intradermal vaccination of C57BL/6 mice with GTL001 adjuvanted with a TLR3 agonist (polyinosinic-polycytidylic acid) or a TLR7 agonist (topical 5% imiquimod cream) induced strong HPV16 E7-specific T-cell responses capable of eradicating HPV16 E7-expressing tumors. Tumor-free mice also had antigen-specific memory T-cell responses that protected them against a subsequent challenge with HPV18 E7-expressing tumor cells. In addition, vaccination with bivalent vaccines containing CyaA-HPV16 E7 and CyaA fused to a tumor-associated antigen (melanoma-specific antigen A3, MAGEA3) or to a non-viral, non-tumor antigen (ovalbumin) eradicated HPV16 E7-expressing tumors and protected against a later challenge with MAGEA3- and ovalbumin-expressing tumor cells, respectively. These results show that CyaA-based bivalent vaccines such as GTL001 can induce both therapeutic and prophylactic anti-tumor T-cell responses. The CyaA platform can be adapted to different antigens and adjuvants, and therefore may be useful for developing other therapeutic vaccines.


Subject(s)
Adenylate Cyclase Toxin/administration & dosage , Drug Carriers/administration & dosage , Immunologic Memory , Papillomavirus Infections/complications , Papillomavirus Vaccines/immunology , T-Lymphocytes/immunology , Uterine Cervical Neoplasms/prevention & control , Adenylate Cyclase Toxin/genetics , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cancer Vaccines/immunology , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Disease Models, Animal , Female , Injections, Intradermal , Melanoma/prevention & control , Mice, Inbred C57BL , Neoplasm Proteins/administration & dosage , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Oncogene Proteins, Viral/administration & dosage , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Ovalbumin/administration & dosage , Ovalbumin/genetics , Ovalbumin/immunology , Ovarian Neoplasms/prevention & control , Papillomavirus E7 Proteins/administration & dosage , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/genetics , Treatment Outcome , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
15.
Int J Mol Med ; 37(2): 436-44, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26676966

ABSTRACT

It is known that vascular smooth muscle cell (VSMC) proliferation and migration leads to intimal hyperplasia in cases of atherosclerosis and restenosis. In the present study, we investigated the effects of kindlin-2 on VSMC proliferation, migration and intimal hyperplasia, and the underlying mechanisms. The left common carotid artery of Sprague­Dawley rats were subjected to balloon injury in order to induce intimal hyperplasia, and then transfected with kindlin-2 small interfering RNA (siRNA) lentivirus or negative control siRNA lentivirus. We noted that the degree of intimal hyperplasia 4 weeks after balloon injury was significantly reduced in arteries transfected with kindlin-2 siRNA lentivirus (P<0.05). In vitro, kindlin-2 siRNA suppressed VSMC proliferation and migration induced by Wnt3a (100 ng/ml). Western blot analyses and RT-qPCR revealed that kindlin-2 regulated Wnt/ß-catenin signaling and thereby modulated the expression of ß-catenin target genes, including c-myc and cyclin D1. This study demonstrated that kindlin-2 plays a critical role in VSMC proliferation, migration and intimal hyperplasia via Wnt signaling. Therefore, blocking the activity of kindlin-2 represents a novel therapeutic strategy for vascular injury.


Subject(s)
Atherosclerosis/therapy , Carotid Arteries/pathology , Hyperplasia/genetics , Membrane Proteins/administration & dosage , Muscle, Smooth, Vascular/pathology , Neoplasm Proteins/administration & dosage , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Carotid Arteries/metabolism , Cell Movement/genetics , Cell Proliferation/genetics , Humans , Hyperplasia/pathology , Hyperplasia/therapy , Membrane Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Neoplasm Proteins/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Rats , Wnt Signaling Pathway/genetics
16.
Fiziol Zh (1994) ; 61(3): 11-8, 2015.
Article in Ukrainian | MEDLINE | ID: mdl-26495731

ABSTRACT

The purpose of this study was to investigate the effect of EMAP II on free radical state of the heart and blood vessels, to restore cNOS coupling and cardiac hemodynamics in spontaneously hypertensive rats. It was found that, due to the combined inhibition of oxidative and nitrosative stress, EMAP I quickly restores impaired in hypertension constitutive de novo synthesis of NO by restoring cNOS coupling. Restoration by EMAP II of constitutive de novo synthesis NO abolished cardiac and endothelial dysfunction in spontaneously hypertensive rats. In hypertension, the introduction of EMAP II helped to improve the performance of the pumping function of the heart (stroke volume increased by 18.2 %, cardiac output -22 %), an arterial stiffness decreased by 23.2 %, process of relaxation of the left ventricle improved, due to decreased in 4,7 times myocardial end-diastolic stiffness.


Subject(s)
Coronary Circulation/drug effects , Cytokines/therapeutic use , Heart/drug effects , Hypertension/drug therapy , Neoplasm Proteins/therapeutic use , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , RNA-Binding Proteins/therapeutic use , Animals , Aorta/metabolism , Cytokines/administration & dosage , Cytokines/pharmacology , Disease Models, Animal , Heart/physiopathology , Heart Function Tests , Humans , Hypertension/enzymology , Hypertension/metabolism , Hypertension/physiopathology , Male , Myocardium/metabolism , Neoplasm Proteins/administration & dosage , Neoplasm Proteins/pharmacology , RNA-Binding Proteins/administration & dosage , RNA-Binding Proteins/pharmacology , Rats, Wistar , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Superoxides/metabolism
17.
J Appl Toxicol ; 35(7): 717-28, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25219328

ABSTRACT

The MAGE-A3 recombinant protein combined with AS15 immunostimulant (MAGE-A3 Cancer Immunotherapeutic) is under development by GlaxoSmithKline for the treatment of lung cancer and melanoma. We performed non-clinical safety studies evaluating potential local and systemic toxic effects induced by MAGE-A3 Cancer Immunotherapeutic in rabbits (study 1) and cynomolgus monkeys (study 2). Animals were allocated to two groups to receive a single (rabbits) or 25 repeated (every 2 weeks) injections (monkeys) of MAGE-A3 Cancer Immunotherapeutic (treatment groups) or saline (control groups). All rabbits were sacrificed 3 days post-injection and monkeys 3 days following last injection (3/5 per gender per group) or after a 3-month treatment-free period (2/5 per gender per group). Local and systemic reactions and MAGE-A3-specific immune responses (monkeys) were assessed. Macroscopic and microscopic (for rabbits, injection site only) post-mortem examinations were performed on all animals. No systemic toxicity or unscheduled mortalities were recorded. Single (rabbits) and repeated (monkeys; up to four times at the same site) injections were well tolerated. Following five to seven repeated injections, limb circumferences increased up to 26% (5 h post-injection), but returned to normal after 1-8 days. Three days after the last injection, enlargements of iliac, popliteal, axillary and inguinal lymph nodes, and increased incidence or severity of mononuclear inflammatory cell infiltrates was observed in injected muscles of treated monkeys. No treatment-related macroscopic findings were recorded after the treatment-free period. MAGE-A3-specific antibody and T-cell responses were raised in all treated monkeys, confirming test item exposure. Single or repeated intramuscular injections of MAGE-A3 Cancer Immunotherapeutic were well tolerated in rabbits and monkeys.


Subject(s)
Antigens, Neoplasm/adverse effects , Neoplasm Proteins/adverse effects , Animals , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/therapeutic use , Drug Administration Schedule , Female , Immunotherapy/methods , Injections, Intramuscular , Macaca fascicularis , Male , Neoplasm Proteins/administration & dosage , Neoplasm Proteins/therapeutic use , Neoplasms/drug therapy , Rabbits , T-Lymphocytes/drug effects , T-Lymphocytes/physiology
19.
Expert Opin Biol Ther ; 14(3): 365-76, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24456101

ABSTRACT

INTRODUCTION: Lung cancer is a common health problem with a bad prognosis, despite recent advances in its treatment. Antigen-specific immunotherapy implies the administration of tumor-specific antigens with an immunostimulant to induce a powerful antitumor immune response, which has shown to be capable of eliminating tumor cells. Melanoma-associated antigen (MAGE) A3 is a good antigen to use in antigen-specific immunotherapy, since it is aberrantly expressed in cancer cells, but not expressed in normal tissue, except in germline and placental cells. AREAS COVERED: Trials have been performed with the MAGE-A3 vaccine in the adjuvant setting after resection of non-small-cell lung cancer. They have shown that the MAGE-A3 vaccine is safe and well tolerated, with promising signs of clinical benefit, especially in patients expressing a specific gene signature. Outcome data are currently expected of a large Phase III randomized controlled trial in the same setting. EXPERT OPINION: The future is hopeful for antigen-specific immunotherapy in general and MAGE-A3 vaccine in specific. Further research needs to identify new tumor-specific antigens, more potent adjuvants and genetic profiles suggestive of a better response toward antigen-specific immunotherapy. The MAGE-A3 vaccine has to be investigated in other settings than the adjuvant one and in other tumor types expressing MAGE-A3.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Neoplasm Proteins/administration & dosage , Vaccination/methods , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Neoplasm/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Humans , Immunotherapy/methods , Lung Neoplasms/immunology , Treatment Outcome , Vaccines, Subunit/immunology
20.
Curr Treat Options Oncol ; 14(4): 580-94, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23934510

ABSTRACT

OPINION STATEMENT: Non-small-cell lung cancer (NSCLC) remains the most common cause of cancer-related death worldwide. Traditional cytotoxic agents and their attendant toxicities have remained the mainstay of systemic therapy for this disease, until now. With the identification of novel molecular and immune cancer-specific aberrancies, molecular agents and immunotherapies have garnered increasing attention as attractive targets, with the potential for improved outcomes while mitigating systemic toxicities seen with traditional cytotoxic agents. Despite a longstanding interest in immunotherapy for the treatment of NSCLC, results of prior studies of therapeutic vaccines have failed to show durable or convincingly meaningful clinical responses. However, newer trials of therapeutic vaccines and checkpoint inhibitors have yielded more promising results. In particular, the checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and the programmed death-1 (PD-1) pathway have shown meaningful clinical responses with manageable toxicities. Large phase III studies are underway, the results of which have the potential to revolutionize the way in which we care for patients with NSCLC. More studies also are needed to investigate the potentially synergistic effects of traditional and immune-based therapies. Given their unique antineoplastic effects, novel immune-specific clinical endpoints also are actively being investigated.


Subject(s)
Antineoplastic Agents/administration & dosage , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Carcinoma, Non-Small-Cell Lung/drug therapy , Immunotherapy , Lung Neoplasms/drug therapy , Molecular Targeted Therapy , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CTLA-4 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Clinical Trials as Topic , Disease-Free Survival , ErbB Receptors/immunology , Female , Humans , Immunotherapy/methods , Ipilimumab , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Male , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/immunology , Neoplasm Proteins/administration & dosage , Neoplasm Proteins/immunology , Nivolumab , Programmed Cell Death 1 Receptor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...