Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Mol Bioeng ; 16(5-6): 431-442, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38099213

ABSTRACT

Purpose: Emergent cancer cells likely secrete factors that inhibit anti-tumor immunity. To identify such factors, we applied a functional assay with proteomics to an immunotherapy resistant syngeneic mouse melanoma model. Four secreted factors were identified that potentially mediate immunosuppression and could become targets for novel immunotherapies. We tested for consistent clinical correlates in existing human data and verified in vivo whether knocking out tumor cell production of these factors improved immune-mediated control of tumor growth. Methods: Existing human data was analyzed for clinical correlates. A CRISPR/Cas9 approach to generate knockout cell lines and a kinetic analysis leveraging a Markov Chain Monte Carlo (MCMC) approach quantified the various knockouts' effect on cells' intrinsic growth rate. Flow cytometry was used to characterize differences in immune infiltration. Results: While all four gene products were produced by malignant melanocytes, only increased CCN4 expression was associated with reduced survival in primary melanoma patients. In immunocompetent C57BL/6 mice the CCN4 knockout increased survival while the other knockouts had no effect. This survival advantage was lost when the CCN4 knockout cells were injected into immunocompromised hosts, indicating that the effect of CCN4 may be immune mediated. Parameter estimation from the MCMC analysis shows that CCN4 was the only knockout tested that decreased the net tumor growth rate in immunocompetent mice. Flow cytometry showed an increase in NK cell infiltration in CCN4 knockout tumors. Conclusions: The results suggest that CCN4 is a mediator of immunosuppression in the melanoma tumor microenvironment and a potential collateral immunotherapy target. Supplementary Information: The online version contains supplementary material available at 10.1007/s12195-023-00787-7.

2.
Nat Commun ; 13(1): 1986, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35418177

ABSTRACT

Developing drugs increasingly relies on mechanistic modeling and simulation. Models that capture causal relations among genetic drivers of oncogenesis, functional plasticity, and host immunity complement wet experiments. Unfortunately, formulating such mechanistic cell-level models currently relies on hand curation, which can bias how data is interpreted or the priority of drug targets. In modeling molecular-level networks, rules and algorithms are employed to limit a priori biases in formulating mechanistic models. Here we combine digital cytometry with Bayesian network inference to generate causal models of cell-level networks linking an increase in gene expression associated with oncogenesis with alterations in stromal and immune cell subsets from bulk transcriptomic datasets. We predict how increased Cell Communication Network factor 4, a secreted matricellular protein, alters the tumor microenvironment using data from patients diagnosed with breast cancer and melanoma. Predictions are then tested using two immunocompetent mouse models for melanoma, which provide consistent experimental results.


Subject(s)
Algorithms , Melanoma , Animals , Bayes Theorem , Carcinogenesis , Gene Regulatory Networks , Humans , Melanoma/genetics , Mice , Transcriptome/genetics , Tumor Microenvironment/genetics
3.
EMBO Rep ; 23(4): e54127, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35099839

ABSTRACT

Cell Communication Network factor 4 (CCN4/WISP1) is a matricellular protein secreted by cancer cells that promotes metastasis by inducing the epithelial-mesenchymal transition. While metastasis limits survival, limited anti-tumor immunity also associates with poor patient outcomes with recent work linking these two clinical correlates. Motivated by increased CCN4 correlating with dampened anti-tumor immunity in primary melanoma, we test for a direct causal link by knocking out CCN4 (CCN4 KO) in the B16F0 and YUMM1.7 mouse melanoma models. Tumor growth is reduced when CCN4 KO melanoma cells are implanted in immunocompetent but not in immunodeficient mice. Correspondingly, CD45+ tumor-infiltrating leukocytes are significantly increased in CCN4 KO tumors, with increased natural killer and CD8+ T cells and reduced myeloid-derived suppressor cells (MDSC). Among mechanisms linked to local immunosuppression, CCN4 suppresses IFN-gamma release by CD8+ T cells and enhances tumor secretion of MDSC-attracting chemokines like CCL2 and CXCL1. Finally, CCN4 KO potentiates the anti-tumor effect of immune checkpoint blockade (ICB) therapy. Overall, our results suggest that CCN4 promotes tumor-induced immunosuppression and is a potential target for therapeutic combinations with ICB.


Subject(s)
Melanoma, Experimental , Melanoma , Animals , CD8-Positive T-Lymphocytes , Cell Communication , Immune Tolerance , Immunosuppression Therapy , Melanoma/metabolism , Mice
4.
Front Aging Neurosci ; 13: 735933, 2021.
Article in English | MEDLINE | ID: mdl-34707491

ABSTRACT

Multiple emerging evidence indicates that the gut microbiota contributes to the pathology of Alzheimer's disease (AD)-a debilitating public health problem in older adults. However, strategies to beneficially modulate gut microbiota and its sensing signaling pathways remain largely unknown. Here, we screened, validated, and established the agonists of free fatty acid receptor 2 (FFAR2) signaling, which senses beneficial signals from short chain fatty acids (SCFAs) produced by microbiota. The abundance of SCFAs, is often low in the gut of older adults with AD. We demonstrated that inhibition of FFAR2 signaling increases amyloid-beta (Aß) stimulated neuronal toxicity. Thus, we screened FFAR2 agonists using an in-silico library of more than 144,000 natural compounds and selected 15 of them based on binding with FFAR2-agonist active sites. Fenchol (a natural compound commonly present in basil) was recognized as a potential FFAR2 stimulator in neuronal cells and demonstrated protective effects against Aß-stimulated neurodegeneration in an FFAR2-dependent manner. In addition, Fenchol reduced AD-like phenotypes, such as Aß-accumulation, and impaired chemotaxis behavior in Caenorhabditis (C.) elegans and mice models, by increasing Aß-clearance via the promotion of proteolysis and reduced senescence in neuronal cells. These results suggest that the inhibition of FFAR2 signaling promotes Aß-induced neurodegeneration, while the activation of FFAR2 by Fenchol ameliorates these abnormalities by promoting proteolytic Aß-clearance and reducing cellular senescence. Thus, stimulation of FFAR2 signaling by Fenchol as a natural compound can be a therapeutic approach to ameliorate AD pathology.

5.
PLoS One ; 15(12): e0243550, 2020.
Article in English | MEDLINE | ID: mdl-33301467

ABSTRACT

HER2/neu is an immunogenic protein inducing both humoral and cell-mediated immune responses. The antigen-specific cytotoxic T lymphocytes (CTLs) are the main effector immune cells in the anti-tumor immunity. To induce an effective CTL specific response against P5+435 single peptide derived from rat HER2/neu oncogene, we used a liposome delivery vehicle. In vivo enhancement of liposome stability and intracytoplasmic delivery of peptides are the main strategies which elevate the liposome-mediated drug delivery. Liposomes containing high transition temperature phospholipids, such as DSPC, are stable with prolonged in vivo circulation and more accessibility to the immune system. Incorporation of DOPE phospholipid results in the effective delivery of peptide into the cytoplasm via the endocytotic pathway. To this end, the P5+435 peptide was linked to Maleimide-PEG2000-DSPE and coupled on the surface of nanoliposomes containing DSPC: DSPG: Cholesterol with/without DOPE. We observed that mice vaccinated with Lip-DOPE-P5+435 formulation had the highest number of IFN-γ- producing CTLs with the highest cytotoxic activity that consequently led to significantly smallest tumor size and prolonged survival rate in the TUBO mice model. In conclusion, our study indicated that the liposomal form of P5+435 peptide containing DOPE can be regarded as a promising prophylactic anti-cancer vaccine to generate potent antigen-specific immunity.


Subject(s)
Breast Neoplasms/prevention & control , Cancer Vaccines/immunology , Receptor, ErbB-2/immunology , Animals , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Drug Delivery Systems/methods , Female , Immunity/drug effects , Interferon-gamma/metabolism , Liposomes/therapeutic use , Mice , Mice, Inbred BALB C , Nanoparticles/therapeutic use , Peptide Fragments/immunology , Peptides/immunology , Pre-Exposure Prophylaxis/methods , T-Lymphocytes, Cytotoxic/immunology
6.
JCI Insight ; 5(9)2020 05 07.
Article in English | MEDLINE | ID: mdl-32302292

ABSTRACT

Inflammation is a major risk factor of morbidity and mortality in older adults. Although its precise etiology is unknown, low-grade inflammation in older adults is commonly associated with increased intestinal epithelial permeability (leaky gut) and abnormal (dysbiotic) gut microbiota. The increasing older population and lack of treatments to reduce aging-related microbiota dysbiosis, leaky gut, and inflammation culminates in a rise in aging-related comorbidities, constituting a significant public health concern. Here, we demonstrate that a human-origin probiotic cocktail containing 5 Lactobacillus and 5 Enterococcus strains isolated from healthy infant gut prevented high-fat diet-induced (HFD-induced) microbiota dysbiosis, leaky gut, inflammation, metabolic dysfunctions, and physical function decline in older mice. Probiotic-modulated gut microbiota primarily reduced leaky gut by increasing tight junctions, which in turn reduced inflammation. Mechanistically, probiotics modulated microbiota in a way to increase bile salt hydrolase activity, which in turn increased taurine abundance in the gut that stimulated tight junctions and suppressed gut leakiness. Furthermore, in Caenorhabditis elegans, taurine increased life span, reduced adiposity and leaky gut, and enhanced physical function. The results suggest that such probiotic therapies could prevent or treat aging-related leaky gut and inflammation in the elderly.


Subject(s)
Aging , Gastrointestinal Microbiome , Inflammation , Probiotics , Tight Junctions , Aging/metabolism , Animals , Caco-2 Cells , Caenorhabditis elegans , Enterococcus/isolation & purification , Humans , Infant , Inflammation/metabolism , Lactobacillus/isolation & purification , Male , Mice , Mice, Inbred C57BL , Probiotics/administration & dosage , THP-1 Cells
7.
J Gerontol A Biol Sci Med Sci ; 75(7): e9-e21, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32129462

ABSTRACT

Aging-related illnesses are increasing and effective strategies to prevent and/or treat them are lacking. This is because of a poor understanding of therapeutic targets. Low-grade inflammation is often higher in older adults and remains a key risk factor of aging-related morbidities and mortalities. Emerging evidence indicates that abnormal (dysbiotic) gut microbiome and dysfunctional gut permeability (leaky gut) are linked with increased inflammation in older adults. However, currently available drugs do not treat aging-related microbiome dysbiosis and leaky gut, and little is known about the cellular and molecular processes that can be targeted to reduce leaky gut in older adults. Here, we demonstrated that metformin, a safe Food and Drug Administration-approved antidiabetic drug, decreased leaky gut and inflammation in high-fat diet-fed older obese mice, by beneficially modulating the gut microbiota. In addition, metformin increased goblet cell mass and mucin production in the obese older gut, thereby decreasing leaky gut and inflammation. Mechanistically, metformin increased the goblet cell differentiation markers by suppressing Wnt signaling. Our results suggest that metformin can be used as a regimen to prevent and treat aging-related leaky gut and inflammation, especially in obese individuals and people with western-style high-fat dietary lifestyle, by beneficially modulating gut microbiome/goblet cell/mucin biology.


Subject(s)
Aging/physiology , Cognition/drug effects , Dysbiosis/prevention & control , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Obesity/complications , Aging/pathology , Aging/psychology , Animals , Diet, High-Fat , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Goblet Cells/drug effects , Inflammation , Mice , Mucins/metabolism , Obesity/metabolism , Obesity/physiopathology , Wnt Signaling Pathway
8.
J Cell Physiol ; 235(5): 4089-4108, 2020 05.
Article in English | MEDLINE | ID: mdl-31642059

ABSTRACT

Unlike other types of breast cancers (BCs), no specific therapeutic targets have been established for triple negative breast cancer (TNBC). Therefore, chemotherapy and radiotherapy are the only available adjuvant therapeutic choices for TNBC. New emerging reports show that TNBC is associated with higher numbers of intratumoral tumor infiltrating lymphocytes. This is indicative of host anti-TNBC immune surveillance and suggesting that immunotherapy can be considered as a therapeutic approach for TNBC management. Recent progress in molecular mechanisms of tumor-immune system interaction and cancer vaccine development studies, fast discoveries and FDA approvals of immune checkpoint inhibitors, chimeric antigen receptor T-cells, and oncolytic virotherapy have significantly attracted attention and research directions toward the immunotherapeutic approach to TNBC. Here in this review different aspects of TNBC immunotherapies including the host immune system-tumor interactions, the tumor microenvironment, the relevant molecular targets for immunotherapy, and clinical trials in the field are discussed.


Subject(s)
Immunotherapy/methods , Peptides/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Female , Humans , Immune Checkpoint Inhibitors/therapeutic use , Tumor Microenvironment
9.
Artif Cells Nanomed Biotechnol ; 47(1): 665-673, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30829072

ABSTRACT

The present study was aimed to develop an effective nanoliposomal vaccine delivery system with P435 HER2/neu-derived peptide conjugated to Maleimide-PEG2000-DSPE. The nanoliposome formulation composed of DSPC/DSPG/Chol/DOPE and monophosphoryl lipid A was used as an adjuvant. Liposomal formulations were prepared and their physical properties were characterized. Anti-tumoral efficacy of formulations was evaluated by immunization of tumor-bearing BALB/c mice and the generated immune response was studied by using ELISpot and flow cytometry analysis. The results of the study demonstrated Lip + DOPE + P535 formulation caused the lowest tumor size and the longest survival time in TUBO mice model and could make it a promising candidate in developing effective vaccines against HER2-positive breast cancers.


Subject(s)
Cancer Vaccines , Mammary Neoplasms, Experimental , Nanoparticles , Peptides , Receptor, ErbB-2 , Animals , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Cell Line, Tumor , Female , Liposomes , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/prevention & control , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Peptides/chemistry , Peptides/immunology , Peptides/pharmacology , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/immunology , Receptor, ErbB-2/therapeutic use
10.
Sci Rep ; 9(1): 2221, 2019 02 18.
Article in English | MEDLINE | ID: mdl-30778090

ABSTRACT

Generating a protective and long-lasting immune response is the primary goal in the expanding field of immunotherapeutic research. In current study we designed an immunogenic bacteriophage- based vaccine to induce a cytotoxic T lymphocyte activity against a mice tumor model over-expressing HER2/neu. Bacteriophage λ displaying a HER2/neu derived peptide GP2 was constructed and used as an anti-cancer vaccine in a BALB/c mouse xenograft tumor model. The results of our study indicated that phage nanoparticles displaying GP2 as a fused peptide to the gpD phage capsid protein induced a robust CTL response. Furthermore, the chimeric phage nanoparticles protected mice against HER2/neu-positive tumor challenge in both prophylactic and therapeutic settings. In conclusion, we propose that λ phage nanoparticles decorated with GP2 peptide merit further investigation for the development of peptide-based vaccines against HER2/neu overexpressing tumors.


Subject(s)
Bacteriophage lambda/genetics , Cancer Vaccines/immunology , Cell Surface Display Techniques , GPI-Linked Proteins/genetics , Nanoparticles , Peptides/genetics , Receptor, ErbB-2/genetics , Animals , Cancer Vaccines/therapeutic use , Cytokines/genetics , Cytokines/metabolism , Cytotoxicity, Immunologic , Disease Models, Animal , Female , GPI-Linked Proteins/immunology , Gene Expression , Humans , Immunization , Mice , Peptide Library , Peptides/immunology , Receptor, ErbB-2/immunology , T-Lymphocytes, Cytotoxic/immunology , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/immunology , Xenograft Model Antitumor Assays
11.
Curr Drug Discov Technol ; 16(3): 251-258, 2019.
Article in English | MEDLINE | ID: mdl-29732989

ABSTRACT

Breast cancer is the second leading cause of cancer death among women. National cancer institute of the US estimates that one in eight women will be diagnosed with breast cancer during their lifetime. Considering the devastating effects of the disease and the alarming numbers many scientists and research groups have devoted their research to fight breast cancer. Several recommendations are to be considered as preventing measures which include living a healthy lifestyle, regular physical activity, weight control and smoking cessation. Early detection of the disease by annual and regular mammography after the age of 40 is recommended by many healthcare institutions. This would help the diagnosis of the disease at an earlier stage and the start of the treatment before it is spread to other parts of the body. Current therapy for breast cancer includes surgical ablation, radiotherapy and chemotherapy which is often associated with adverse effects and even may lead to a relapse of the disease at a later stage. In order to achieve a long-lasting anticancer response with minimal adverse effects, development of breast cancer vaccines is under investigation by many laboratories. The immune system can be stimulated by a vaccine against breast cancer. This approach has attracted a great enthusiasm in recent years. No breast cancer vaccines have been approved for clinical use today. One breast cancer vaccine (NeuVax) has now completed clinical trial phase III and a few preventive and therapeutic breast cancer vaccines are at different steps of development. We think that with the recent advancements in immunotherapy, a breast cancer vaccine is not far from reach.


Subject(s)
Breast Neoplasms/therapy , Cancer Vaccines , Immunotherapy , Animals , Female , Humans
12.
Cancer Lett ; 424: 109-116, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29580807

ABSTRACT

Phage display technique has been increasingly researched for vaccine design and delivery strategies in recent years. In this study, the AE37 (Ii-Key/HER-2/neu 776-790) peptide derived from HER2 (human epidermal growth factor receptor protein) was used as a fused peptide to the lambda phage (λF7) coat protein gpD, and the phage nanoparticles were used to induce antitumor immunogenicity in a TUBO model of breast cancer in mice. Mice were immunized with the AE37 peptide displaying phage, λF7 (gpD::AE37) every 2-week intervals over 6-weeks, then the generated immune responses were evaluated. An induction of CTL immune response by the λF7 (gpD::AE37) construct compared to the control λF7 and buffer groups was observed in vitro. Moreover, in the in vivo studies, the vaccine candidate showed promising prophylactic and therapeutic effects against the HER2 overexpressing cancer in BALB/c mice.


Subject(s)
Bacteriophage lambda/genetics , Breast Neoplasms/drug therapy , Cancer Vaccines/administration & dosage , Capsid Proteins/genetics , Glycoproteins/genetics , Recombinant Fusion Proteins/administration & dosage , Animals , Bacteriophage lambda/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Capsid Proteins/immunology , Cell Line, Tumor , Female , Glycoproteins/immunology , Humans , Immunization , Mice , Mice, Inbred BALB C , Nanoparticles , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Receptor, ErbB-2/genetics , Recombinant Fusion Proteins/immunology , Treatment Outcome , Xenograft Model Antitumor Assays
13.
J Drug Target ; 26(4): 365-372, 2018 04.
Article in English | MEDLINE | ID: mdl-28972792

ABSTRACT

E75 (HER-2/neu-369-377), is an immunogenic peptide which is highly expressed in breast cancer patients. The purpose of this study was to develop an effective vaccine delivery/adjuvant system by attachment of this peptide to the surface of liposomes consisting of phospholipids including distearoylphosphocholine (DSPC) and distearoyl phosphoglycerol (DSPG) with high transition temperature (Tm) and dioleoylphosphatidylethanolamine (DOPE) (a pH-sensitive lipid for cytosolic antigen delivery) to improve antitumour immune activity against the E75 peptide. For this purpose, the E75 peptide was incorporated into liposomes consisting of DSPC/DSPG/cholesterol (Chol)/DOPE (15/2/3/5 molar ratio) through conjugation with distearoylphosphoethanolamine-N-[maleimide(polyethylene glycol)-2000] (maleimide-PEG2000-DSPE). Immunization of BALB/c mice was performed three times with different forms of liposomal formulations at 2-week intervals and antitumour immunity responses were evaluated. Results of ELISpot and flow cytometry analysis showed that mice vaccinated with DSPC/DSPG/Chol/DOPE/E75 have significantly enhanced the antigen-specific IFN-γ response of CD8+ T cells and generated cytotoxic T lymphocytes (CTL) antitumour responses. CTL responses induced by this formulation resulted in inhibition of tumour progression and longer survival time in the mice TUBO tumour model. The results revealed that the liposomes consist of DSPC/DSPG/Chol/DOPE could be suitable candidates for vaccine delivery of E75 peptide for the prevention and therapy of HER2-positive breast cancer and merit further investigation.


Subject(s)
Breast Neoplasms/drug therapy , Cancer Vaccines/administration & dosage , Peptide Fragments/administration & dosage , Phospholipids/chemistry , Receptor, ErbB-2/administration & dosage , Animals , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Disease Progression , Female , Interferon-gamma/immunology , Liposomes , Mice , Mice, Inbred BALB C , Nanoparticles , Peptide Fragments/immunology , Receptor, ErbB-2/immunology , Survival Rate , T-Lymphocytes, Cytotoxic/immunology , Transition Temperature
14.
Immunol Res ; 66(1): 200-206, 2018 02.
Article in English | MEDLINE | ID: mdl-29143917

ABSTRACT

We have investigated the in vitro immunogenicity and in vivo prophylactic and therapeutic potential of lambda (λ) phage particles displaying the E75 peptide (derived from HER2 protein) in an implantable TUBO breast tumor model of BALB/c mice. The mice were immunized with the E75-displaying phage (λF7-gpD::E75) every 2-week intervals over a 6-week period, and the generated immune responses were studied. Results showed in vitro induction of immune responses by the λF7 (gpD::E75) construct compared to the control λF7 and buffer groups. In the in vivo prophylactic study, all the control and vaccinated mice groups developed tumors. However, in the therapeutic experiments, we observed a significant difference in tumor size at days 14-36 for mice immunized with λF7 (gpD::E75) compared to control groups (P < 0.05). Moreover, the survival time prolonged in mice immunized with λF7 (gpD::E75). The discrepancy between the results obtained from the in vitro and in vivo studies may have been a result of the induction of Foxp3 CD4+CD25+ which has been previously reported to hamper effective T cell functionality. In conclusion, we observed a significant immune stimulatory response in the in vitro study, while in vivo, the vaccine was not able to exert significant tumor inhibitory effects. We suggest that the presence of Foxp3+ CD4+CD25+ cells may have impaired the anti-tumor response in mice challenged in vivo with the TUBO xenograft tumor.


Subject(s)
Bacteriophage lambda/physiology , Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Glucosephosphate Dehydrogenase/immunology , Peptide Fragments/immunology , Receptor, ErbB-2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Disease Models, Animal , Female , Forkhead Transcription Factors/metabolism , Glucosephosphate Dehydrogenase/genetics , Humans , Mice , Mice, Inbred BALB C , Nanoparticles , Peptide Fragments/genetics , Receptor, ErbB-2/genetics , Tumor Burden , Vaccination
15.
PLoS One ; 12(10): e0185099, 2017.
Article in English | MEDLINE | ID: mdl-29045460

ABSTRACT

One of the challenging issues in vaccine development is peptide and adjuvant delivery into target cells. In this study, we developed a vaccine and therapeutic delivery system to increase cytotoxic T lymphocyte (CTL) response against a breast cancer model overexpressing HER2/neu. Gp2, a HER2/neu-derived peptide, was conjugated to Maleimide-mPEG2000-DSPE micelles and post inserted into liposomes composed of DMPC, DMPG phospholipids, and fusogenic lipid dioleoylphosphatidylethanolamine (DOPE) containing monophosphoryl lipid A (MPL) adjuvant (DMPC-DMPG-DOPE-MPL-Gp2). BALB/c mice were immunized with different formulations and the immune response was evaluated in vitro and in vivo. ELISpot and intracellular cytokine analysis by flow cytometry showed that the mice vaccinated with Lip-DOPE-MPL-GP2 incited the highest number of IFN-γ+ in CD8+ cells and CTL response. The immunization led to lower tumor sizes and longer survival time compared to the other groups of mice immunized and treated with the Lip-DOPE-MPL-GP2 formulation in both prophylactic and therapeutic experiments. These results showed that co-formulation of DOPE and MPL conjugated with GP2 peptide not only induces high antitumor immunity but also enhances therapeutic efficacy in TUBO mice model. Lip-DOPE-MPL-GP2 formulation could be a promising vaccine and a therapeutic delivery system against HER2 positive cancers and merits further investigation.


Subject(s)
Breast Neoplasms/immunology , Cancer Vaccines/immunology , Liposomes/chemistry , Nanoparticles/chemistry , Peptides/immunology , Receptor, ErbB-2/metabolism , Xenograft Model Antitumor Assays , Amino Acid Sequence , Animals , Antigens, Neoplasm/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Separation , Chromatography, Thin Layer , Cytokines/metabolism , Enzyme-Linked Immunospot Assay , Female , Interferon-gamma/metabolism , Interleukin-4/metabolism , Lymph Nodes/metabolism , Maleimides/chemistry , Mice, Inbred BALB C , Peptides/chemistry , Phenotype , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Real-Time Polymerase Chain Reaction , Spleen/pathology , T-Lymphocytes, Cytotoxic/immunology , Vaccination
16.
Immunol Lett ; 190: 108-117, 2017 10.
Article in English | MEDLINE | ID: mdl-28736158

ABSTRACT

This study was designed to prepare and characterize nanoliposomal vaccine formulation encapsulating AE36 HER2/neu-derived peptide with or without CpG and evaluate the immunologic and therapeutic responses of that in BALB/c mice model of Her2 overexpressing breast cancer. AE36 was encapsulated in liposomes composed of DOTAP, DOPE and Cholesterol (DDC) or DD with. The formulations could induce both CD8+ and CD4+ responses and stimulate production of cytokines which was detected by Enzyme-linked immunospot assay (ELISpot) kits, cytotoxicity test and intracellular cytokine assay by flow cytometry. The formulation showed both therapeutic and prophylactic effects in BALB/c mice bearing Her2+ breast cancer. DDC+CpG showed the best effect in prophylactic study and DD+pG showed the best effect in therapeutic study, which both of them decreased the size of tumors significantly. The engineered nanoliposomes containing AE36 could be a candidate vaccine for the treatment or prophylaxis of HER2+ breast cancer and merits further investigation.


Subject(s)
Breast Neoplasms/therapy , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Immunotherapy/methods , Liposomes/therapeutic use , Nanostructures/statistics & numerical data , Peptides/therapeutic use , Receptor, ErbB-2/therapeutic use , Animals , Breast Neoplasms/immunology , Cell Line, Tumor , Cholesterol/chemistry , Cytokines/metabolism , Cytotoxicity, Immunologic , Disease Models, Animal , Fatty Acids, Monounsaturated/chemistry , Female , Humans , Liposomes/chemistry , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/administration & dosage , Phosphatidylethanolamines/chemistry , Quaternary Ammonium Compounds/chemistry
17.
Cell Biol Int ; 37(4): 348-58, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23450781

ABSTRACT

The broad spectrum of the pharmacological effects of sulphonamide family of drugs motivated us to investigate the cellular mechanisms for anti-cancer effects of sulphathiazole and sulphacetamide on T-47D breast cancer cells. Fluorescent microscopy, flow cytometric analysis, caspase-3 activity and DNA fragmentation assays were used to detect apoptosis. The distribution of the cells among different phases of the cell cycle was measured by flow cytometry. The expression of several genes with important roles in some critical cellular pathways including apoptosis, mTOR/AKT pathway and autophagy were determined by real-time RT-PCR analysis. Sulphathiazole and sulphacetamide induced anti-proliferative effects on T-47D cells were independent of apoptosis and cell cycle arrest. The overexpression of critical genes involved in autophagy including ATG5, p53 and DRAM indicated that the main effect of the drug-induced anti-proliferative effects was through induction of autophagy. This process was induced in two different forms, including death inducing and cytoprotective autophagy. Sulphathiazole treatment was followed by higher expression of p53/DRAM and downregulation of Akt/mTOR pathway resulting in death autophagy. In contrast, sulphacetamide treatment lowered expression of p53/DRAM pathway in parallel with upregulation of Akt/mTOR pathway promoting cytoprotective autophagy. The results indicated that autophagy is the main mechanism mediating the anti-cancer effects of sulphathiazole and sulphacetamide on T-47D cells. Alignment of the p53 and DRAM expression along with activation level of Akt survival pathway therefore determines the type of autophagy that occurs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Autophagy/drug effects , Sulfacetamide/pharmacology , Sulfathiazoles/pharmacology , Apoptosis , Caspase 3/metabolism , Cell Cycle , Cell Line, Tumor , Cytoprotection , DNA Fragmentation , Enzyme Activation , Humans , Lethal Dose 50 , Sulfathiazole
SELECTION OF CITATIONS
SEARCH DETAIL
...