Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 98
1.
Technol Cancer Res Treat ; 20: 15330338211034260, 2021.
Article En | MEDLINE | ID: mdl-34318732

PURPOSE: Human umbilical endothelial cells (HUVECs) have been proved to be an effective whole-cell vaccine inhibiting tumor angiogenesis. In this study, we fused HUVECs with human lung adenocarcinoma cells A549 s, aiming at preparing lung cancer vaccine to achieve dual effects of anti-tumor angiogenesis and specific immunity to tumor cells. METHODS: A549 cells were induced by ethyl methane sulfonate (EMS) and 8-azaguanine (8-AG) to get hypoxanthine guanine phosphoribosyl transferase (HGPRT) auxotrophic A549 cells. Then Fused HGPRT auxotrophic A549 cells with primary HUVEC cells by combining electrofusion with polyethylene glycol (PEG). Afterward the fusion cells were screened by HAT and HT selective medium and sorted by flow cell sorter to obtain high-purity HUVEC-A549 cells. Finally, HUVEC-A549 cells were identified by karyotype analysis and western blotting. RESULTS: The fusion efficiency of HUVEC-A549 cells prepared by combining electrofusion with polyethylene glycol (PEG) was significantly higher than that of electrofusion and PEG (43.0% vs 17.60% vs 2.71%, P < 0.05). After screened by HAT and HT selective medium and sorted by flow cell sorter, the proportion of HUVEC-A549 cells can count for 71.2% ± 3.2%. The mode of chromosomes in HUVEC-A549 cells was 68, and the chromosome was triploid. VE-cadherin and platelet endothelial cell adhesion molecule-1 (CD31) were highly expressed in HUVECs and HUVEC-A549 cells, but not in A549 cells. CONCLUSIONS: These results indicate that HUVEC-A549 cells retain the biological characteristics of human umbilical vein endothelial cells and A549 cells. It can be used in the experimental study of lung cancer cell vaccine.


Cancer Vaccines/biosynthesis , Carcinoma, Non-Small-Cell Lung/therapy , Cell Fusion/methods , Lung Neoplasms/therapy , A549 Cells , Human Umbilical Vein Endothelial Cells , Humans , Immunotherapy , Karyotype , Neovascularization, Pathologic/therapy , Polyethylene Glycols
2.
Eur J Surg Oncol ; 47(9): 2460-2464, 2021 09.
Article En | MEDLINE | ID: mdl-33980416

INTRODUCTION: The induction of adaptive cellular immunity in patients with in-transit melanoma metastasis treated with hyperthermic isolated limb perfusion (ILP) with melphalan has been shown to contribute to the effectiveness of the therapy. Activated CD8+ T cells appear to be of particular importance for the efficacy of melphalan-based ILP therapy, as observed in both patients and animal models. In this study, we explored the possible synergistic effects of combining melphalan-based therapy with the checkpoint inhibitor anti-PD-1 on tumours in a mouse melanoma model. METHODS: A murine vaccination model that utilized melphalan-exposed melanoma cells was used to mimic certain immunological features of melphalan-based ILP. The effects of the vaccine on tumour growth and PD-1 expression on CD8+ tumour-infiltrating T cells were analyzed. The melphalan-based vaccine was then combined with an anti-PD-1 antibody and tumour growth was assessed. RESULTS: Treatment with melphalan-based therapy significantly induced the expression of PD-1 on CD8+ tumour-infiltrating lymphocytes. Combination therapy using melphalan-based therapy followed by treatment with PD-1 antibodies significantly reduced early-stage tumour growth relative to monotherapies and no treatment. CONCLUSIONS: This study thus suggests that the addition of PD-1 blockade to melphalan-based therapies, such as ILP, may be therapeutically beneficial.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Immunotherapy/methods , Melanoma, Experimental/drug therapy , Programmed Cell Death 1 Receptor/metabolism , Skin Neoplasms/drug therapy , Animals , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/biosynthesis , Female , Immune Checkpoint Inhibitors/administration & dosage , Lymphocytes, Tumor-Infiltrating/metabolism , Melphalan/administration & dosage , Mice , Mice, Inbred C57BL
3.
Curr Protein Pept Sci ; 22(1): 74-88, 2021.
Article En | MEDLINE | ID: mdl-33208071

Breast cancer is a heterogeneous malignancy and is the second leading cause of mortality among women around the world. Increasing the resistance to anti-cancer drugs in breast cancer cells persuades researchers to search the novel therapeutic approaches for the treatment of this malignancy. Among the novel methods, therapeutic peptides that target and disrupt tumor cells have been of great interest. Therapeutic peptides are short amino acid monomer chains with high specificity to bind and modulate a protein interaction of interest. Several advantages of peptides, such as specific binding on tumor cells surface, low molecular weight, and low toxicity on normal cells, make the peptides appealing therapeutic agents against solid tumors, particularly breast cancer. Also, the National Institutes of Health (NIH) describes therapeutic peptides as a suitable candidate for the treatment of drug-resistant breast cancer. In this review, we attempt to review the different therapeutic peptides against breast cancer cells that can be used in the treatment and diagnosis of the malignancy. Meanwhile, we presented an overview of peptide vaccines that have been developed for the treatment of breast cancer.


Antineoplastic Agents/therapeutic use , BRCA1 Protein/genetics , Breast Neoplasms/drug therapy , Cancer Vaccines/therapeutic use , Cell-Penetrating Peptides/therapeutic use , Neoplasm Proteins/genetics , Antineoplastic Agents/chemical synthesis , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cancer Vaccines/biosynthesis , Cancer Vaccines/immunology , Cell Death/drug effects , Cell Death/genetics , Cell-Penetrating Peptides/biosynthesis , Cell-Penetrating Peptides/chemical synthesis , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Tumor Cells, Cultured , Vaccines, Subunit
4.
Br J Cancer ; 123(9): 1387-1394, 2020 10.
Article En | MEDLINE | ID: mdl-32753678

BACKGROUND: Peptide-vaccination therapy targeting tumour-associated antigens can elicit immune responses, but cannot be used to eliminate large tumour burden. In this study, we developed a therapeutic single-chain variable-fragment (scFv) antibody that recognises the cancer stem-like cell/cancer-initiating cell (CSC/CIC) antigen, DNAJB8. METHODS: We screened scFv clones reacting with HLA-A24:20/DNAJB8-derived peptide (DNAJB8_143) complex using naive scFv phage-display libraries. Reactivity and affinity of scFv clones against the cognate antigen were quantified using FACS and surface plasmon resonance. Candidate scFv clones were engineered to human IgG1 (hIgG1) and T-cell-engaging bispecific antibody (CD3xJB8). Complement-dependent cytotoxicity (CDC) and bispecific antibody-dependent cellular cytotoxicity (BADCC) were assessed. RESULTS: scFv clones A10 and B10 were isolated after bio-panning. Both A10-hIgG1 and B10-hIgG1 reacted with DNAJB8-143 peptide-pulsed antigen-presenting cells and HLA-A24(+)/DNAJB8(+) renal cell carcinoma and osteosarcoma cell lines. A10-hIgG1 and B10-hIgG1 showed strong affinity with the cognate HLA/peptide complex (KD = 2.96 × 10-9 M and 5.04 × 10-9 M, respectively). A10-hIgG1 and B10-hIgG1 showed CDC against HLA-A24(+)/DNAJB8(+) cell lines. B10-(CD3xJB8) showed superior BADCC to A10-(CD3xJB8). CONCLUSION: We isolated artificial scFv antibodies reactive to CSC/CIC antigen DNAJB8-derived peptide naturally present on renal cell carcinoma and sarcoma. Immunotherapy using these engineered antibodies could be promising.


HLA-A24 Antigen/immunology , HSP40 Heat-Shock Proteins/immunology , Immunotherapy/methods , Molecular Chaperones/immunology , Neoplastic Stem Cells/immunology , Nerve Tissue Proteins/immunology , Protein Engineering/methods , Single-Chain Antibodies/biosynthesis , Antibody Specificity , Antibody-Dependent Cell Cytotoxicity , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Cancer Vaccines/biosynthesis , Cancer Vaccines/therapeutic use , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , HEK293 Cells , HLA-A24 Antigen/genetics , HLA-A24 Antigen/metabolism , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , HT29 Cells , Humans , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Osteosarcoma/immunology , Osteosarcoma/pathology , Osteosarcoma/therapy , Peptide Fragments/immunology , Peptide Fragments/metabolism , Peptide Library , Single-Chain Antibodies/therapeutic use
5.
Protein Expr Purif ; 174: 105658, 2020 10.
Article En | MEDLINE | ID: mdl-32360598

The recombinant multi-epitope vaccine called VBP3 is designed to suppress tumor growth and angiogenesis through targeting both basic fibroblast growth factor (bFGF) and vascular endothelial growth factor A (VEGFA). We are aiming to produce VBP3 vaccine in a large scale and provide sufficient protein for pre-clinical study. High cost and potential toxicity are severe limitations of IPTG and we investigated whether lactose can mediate VBP3 induction. Firstly, we identified the biological characteristics and established a culture bank of VBP3 strains. The best-performing strains were selected and the fermentation mode of medium, bacterial growth and protein expression were optimized in shake flasks. We scaled up the VBP3 production in 10 L bioreactor using lactose as inducer and the protein yield was comparable with IPTG induction. Next, the target protein was purified by nickel-nitrilotriacetic acid (Ni-NTA) affinity chromatography, with a SDS-PAGE purity over 90%. Further, the purified VBP3 vaccine was subcutaneously injected in BALB/c mice and elicited high-titer anti-bFGF (1:32,000) and anti-VEGFA (1:4000) antibodies. Take together, lactose was an applicable inducer for VBP3 production and the eligible product of VBP3 was harvested in the large-scale fermentation, supporting the industrial production and pre-clinical study in the future. The VBP3 vaccine with superior immunogenicity might be used as a potential therapeutic vaccine for tumor treatment.


Cancer Vaccines , Escherichia coli/growth & development , Fibroblast Growth Factor 2 , Recombinant Fusion Proteins , Vascular Endothelial Growth Factor A , Animals , Cancer Vaccines/biosynthesis , Cancer Vaccines/genetics , Cancer Vaccines/isolation & purification , Escherichia coli/genetics , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/isolation & purification , Fibroblast Growth Factor 2/pharmacology , Humans , Immunogenicity, Vaccine , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/isolation & purification , Vascular Endothelial Growth Factor A/pharmacology
6.
Biochem Biophys Res Commun ; 524(4): 825-831, 2020 04 16.
Article En | MEDLINE | ID: mdl-32037086

Chromatin organization starts from a "beads-on-a string" 10 nm fiber, a basic nucleosomal structure consisting of DNA and core histones. Given its regular nucleosome array on DNA backbone where N-terminal tails of each histone are exposed on the surface of chromatin fiber, we hypothesized that chromatin can be utilized as a heterologous peptide carrier to elicit a peptide-specific immune response. The plasmid DNA containing the Widom's clone 601 sequence and the recombinant chimeric histones containing the peptide derived from ras oncogene (G12V) were used to assemble the chromatin fiber in vitro. The immunogenicity of the assembled chromatin was tested in mice as a single vaccine component or formulated with adjuvants. G12V tagged-chromatin co-administered with adjuvants induced higher antibody responses against the G12V peptide than vaccination with adjuvant alone, while chimeric histones did not generate a significant antibody response. Interestingly, splenocytes from mice vaccinated with the G12V tagged-chromatin vaccine did not generate significant antigen-specific cytokine responses. Our studies suggest that chromatin can be utilized as an effective carrier of antigenic peptides for inducing specific antibody responses.


Cancer Vaccines/biosynthesis , Genes, ras/immunology , Histones/immunology , Nanofibers/chemistry , Peptide Library , Peptides/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Antibodies/genetics , Antibodies/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Chromatin Assembly and Disassembly , Histones/genetics , Histones/metabolism , Humans , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/prevention & control , Nucleosomes/chemistry , Nucleosomes/immunology , Nucleosomes/metabolism , Peptides/genetics , Peptides/metabolism , Plasmids/chemistry , Plasmids/immunology , Plasmids/metabolism , Vaccines, Subunit , Xenopus laevis
9.
Immunity ; 47(6): 1037-1050.e6, 2017 12 19.
Article En | MEDLINE | ID: mdl-29221729

Given the limited efficacy of clinical approaches that rely on ex vivo generated dendritic cells (DCs), it is imperative to design strategies that harness specialized DC subsets in situ. This requires delineating the expression of surface markers by DC subsets among individuals and tissues. Here, we performed a multiparametric phenotypic characterization and unbiased analysis of human DC subsets in blood, tonsil, spleen, and skin. We uncovered previously unreported phenotypic heterogeneity of human cDC2s among individuals, including variable expression of functional receptors such as CD172a. We found marked differences in DC subsets localized in blood and lymphoid tissues versus skin, and a striking absence of the newly discovered Axl+ DCs in the skin. Finally, we evaluated the capacity of anti-receptor monoclonal antibodies to deliver vaccine components to skin DC subsets. These results offer a promising path for developing DC subset-specific immunotherapies that cannot be provided by transcriptomic analysis alone.


Antigens, Differentiation/immunology , Biological Variation, Individual , Dendritic Cells/immunology , Phenotype , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Immunologic/immunology , Skin/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation/genetics , Biomarkers/analysis , Cancer Vaccines/administration & dosage , Cancer Vaccines/biosynthesis , Cytophotometry/methods , Dendritic Cells/cytology , Female , Gene Expression , Humans , Immunophenotyping , Immunotherapy , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Organ Specificity , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Immunologic/genetics , Skin/cytology , Spleen/cytology , Spleen/immunology , Axl Receptor Tyrosine Kinase
11.
Protein Expr Purif ; 134: 132-138, 2017 Jun.
Article En | MEDLINE | ID: mdl-28410993

LHRH based vaccines are promising candidates for therapy of androgen and estrogen dependent cancers. We report in this communication development of a novel recombinant protein vaccine candidate against LHRH. A synthetic gene was designed in which the codon sequence in the LHRH decapeptide was modified by substituting the codon for 6-glycine with that of l-leucine. Further the LHRH(6leu) gene was linked to heat-labile enterotoxin of E. coli (LTB) as carrier. This LHRH(6leu)-LTB gene was cloned into a prokaryotic expression vector under the control of inducible and strong bacteriophage T7 promoter to over-express LHRH(leu) fused to LTB as recombinant protein in E. coli. Recombinant LHRH(leu)-LTB protein of ∼14 kDa size, was purified from inclusion bodies using in-situ refolding on the column and Ni-NTA based immobilized affinity chromatography. Western blot confirmed the immunoreactivity of purified LHRH(leu)-LTB fusion protein with anti-LHRH monoclonal antibody. The vaccine protein was further characterized by mass spectroscopy, circular dichroism and fluorescence spectroscopy. This communication reports a recombinant LHRH fusion protein with potential for blocking of sex hormones production for eventual therapy of sex hormones dependent neoplasms.


Androgens , Cancer Vaccines , Estrogens , Gonadotropin-Releasing Hormone , Lymphotoxin-beta , Neoplasms/therapy , Recombinant Fusion Proteins , Cancer Vaccines/biosynthesis , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Gonadotropin-Releasing Hormone/biosynthesis , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/therapeutic use , Humans , Lymphotoxin-beta/biosynthesis , Lymphotoxin-beta/therapeutic use , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use
12.
Virus Res ; 233: 35-41, 2017 04 02.
Article En | MEDLINE | ID: mdl-28286036

In order to test and enhance the antitumor activity against mice melanoma by NDV-modified tumor vaccine, a recombinant NDV expressing IL-15 (LX/(IL-15)) was generated by reverse genetics. Then, the expression level and biological activity of IL-15 were examined. Our results showed that mice tumor cell lines infected with LX/(IL-15) expressed IL-15 at a high level, and that expressed IL-15 was biologically active. Expression kinetics demonstrated that the highest expression level of IL-15 was at 48h post infection. The cytotoxicity assay showed that murine melanoma cells modified with LX/(IL-15) could significantly enhance the antitumor immune response in vitro. In vivo study also showed that murine melanoma cells modified with LX/(IL-15) could prevent melanoma growth in mice. Taken together, our data strongly indicated that recombinant LX/(IL-15) is a promising agent for cancer immunotherapy both for human and animal.


Cancer Vaccines/biosynthesis , Interleukin-15/immunology , Melanoma, Experimental/therapy , Newcastle disease virus/immunology , Oncolytic Viruses/immunology , Skin Neoplasms/therapy , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Coculture Techniques , Cytotoxicity, Immunologic/genetics , Female , Gene Expression , Humans , Immunotherapy/methods , Interleukin-15/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Newcastle disease virus/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Reverse Genetics/methods , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Transgenes
13.
Sci Rep ; 7: 42145, 2017 02 13.
Article En | MEDLINE | ID: mdl-28191816

Active human dendritic cells (DCs), which efficiently induce immune responses through their functions as antigen-presenting cells, exhibit direct anti-tumour killing activity in response to some pathogens and cytokines. These antigen-presenting and tumour killing abilities may provide a breakthrough in cancer immunotherapy. However, the mechanisms underlying this killer DC activity have not been fully proven, despite the establishment of interferon-α (IFN-α)-generated killer DCs (IFN-DCs). Here mature IFN-DCs (mIFN-DCs), generated from IFN-DCs primed with OK-432 (streptococcal preparation), exhibited elevated expression of CD86 and human leukocyte antigen-DR (minimum criteria for DC vaccine clinical trials) as well as antigen-presenting abilities comparable with those of mature IL-4-DCs (mIL-4-DCs). Interestingly, the killing activity of mIFN-DCs, which correlated with the expression of CD56 (natural killer cell marker) and was activated via the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and Fas ligand pathway, was stronger than that of IFN-DCs and remarkably stronger than that of mIL-4-DCs. Therefore, mIFN-DCs exhibit great potential as an anti-cancer vaccine that would promote both acquired immunity and direct tumour killing.


Cytotoxicity, Immunologic , Dendritic Cells/drug effects , Fas Ligand Protein/genetics , Interferon-alpha/pharmacology , Interleukin-4/pharmacology , Picibanil/pharmacology , TNF-Related Apoptosis-Inducing Ligand/genetics , Antigen Presentation , B7-2 Antigen/genetics , B7-2 Antigen/immunology , CD56 Antigen/genetics , CD56 Antigen/immunology , Cancer Vaccines/biosynthesis , Cell Differentiation/drug effects , Dendritic Cells/cytology , Dendritic Cells/immunology , Dinoprostone/pharmacology , Fas Ligand Protein/immunology , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Humans , Immunophenotyping , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Primary Cell Culture , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/immunology
14.
Vaccine ; 35(7): 1094-1100, 2017 02 15.
Article En | MEDLINE | ID: mdl-27449681

The development of next generation sequencing technologies has revolutionized our understanding of how specific genetic events contribute to cancer initiation and progression. Dramatic improvements in instrument design and efficiency, combined with significant cost reductions has permitted a systematic analysis of the mutational landscape in a variety of cancer types. At the same time, a detailed map of the cancer mutanome in individual cancers offers a unique opportunity to develop personalized cancer vaccine strategies targeting neoantigens. Recent studies in both preclinical models and human cancer patients demonstrate that neoantigens (1) are important targets following checkpoint inhibition therapy, (2) have been identified as the target of adoptive T cell therapies, and (3) can be successfully targeted with personalized vaccines. Taken together, these observations provide strong rationale for the clinical translation of personalized cancer vaccines.


Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Molecular Targeted Therapy , Mutation , Neoplasms/therapy , Precision Medicine/methods , Antigen Presentation , Antigens, Neoplasm/genetics , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/biosynthesis , Cancer Vaccines/genetics , Clinical Trials as Topic , Genetic Vectors/chemistry , Genetic Vectors/metabolism , High-Throughput Nucleotide Sequencing , Humans , Immunotherapy/methods , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Peptides/chemical synthesis , Peptides/immunology , Peptides/therapeutic use , Precision Medicine/instrumentation , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Vaccines, DNA/biosynthesis , Vaccines, DNA/genetics , Vaccines, DNA/therapeutic use
15.
Virus Res ; 231: 148-165, 2017 03 02.
Article En | MEDLINE | ID: mdl-27932207

Human papillomavirus (HPV) is known to be a necessary factor for many gynecologic malignancies and is also associated with a subset of head and neck malignancies. This knowledge has created the opportunity to control these HPV-associated cancers through vaccination. However, despite the availability of prophylactic HPV vaccines, HPV infections remain extremely common worldwide. In addition, while prophylactic HPV vaccines have been effective in preventing infection, they are ineffective at clearing pre-existing HPV infections. Thus, there is an urgent need for therapeutic and T cell-based vaccines to treat existing HPV infections and HPV-associated lesions and cancers. Unlike prophylactic vaccines, which generate neutralizing antibodies, therapeutic, and T cell-based vaccines enhance cell-mediated immunity against HPV antigens. Our review will cover various therapeutic and T cell-based vaccines in development for the treatment of HPV-associated diseases. Furthermore, we review the strategies to enhance the efficacy of therapeutic vaccines and the latest clinical trials on therapeutic and T cell-based HPV vaccines.


Dendritic Cells/transplantation , Head and Neck Neoplasms/prevention & control , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , T-Lymphocytes/transplantation , Uterine Cervical Neoplasms/prevention & control , Vaccination , Adoptive Transfer , Cancer Vaccines/administration & dosage , Cancer Vaccines/biosynthesis , Cancer Vaccines/immunology , Clinical Trials as Topic , Dendritic Cells/immunology , Female , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/virology , Humans , Immunity, Cellular/drug effects , Papillomaviridae/drug effects , Papillomaviridae/growth & development , Papillomaviridae/immunology , Papillomaviridae/pathogenicity , Papillomavirus Infections/immunology , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/biosynthesis , T-Lymphocytes/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Vaccines, DNA/administration & dosage , Vaccines, DNA/biosynthesis , Vaccines, DNA/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/immunology
16.
Recent Pat Biotechnol ; 11(1): 32-41, 2017.
Article En | MEDLINE | ID: mdl-27903220

BACKGROUND: Development of a universal cancer vaccine for the prevention of all cancers has been under development for many years. Antiangiogenic cancer vaccines elicit immune responses with the potential of destroying tumor vasculature endothelial cells without affecting vasculature integrity in normal tissues. The methods used in the development of antigen compositions comprising these vaccines have been recently improved and described in this report in the context of SANTAVAC ™ development - the first cancer vaccine based on endothelial cell heterogeneity. METHODS: The present report summarizes data related to SANTAVAC™ development, including technical key points associated with optimal SANTAVAC™ production, a description of the composition required for preparing cancer vaccines with the highest predicted efficacy and safety, and a strategy for SANTAVAC™ large-scale implementation. Patents related to SANTAVAC™ and other universal cancer vaccines are also described. RESULTS: SANTAVAC ™ was shown to be the most promising antigen composition for anti-cancer vaccination, allowing for immune targeting of the tumor vasculature in experimental models with a high predicted efficacy (up to 60), where efficacy represents the fold decrease in the number of endothelial cells with a tumor-induced phenotype and directly related to predicted arrest of tumor growth. CONCLUSION: The use of SANTAVAC ™ as a universal antigenic composition may spur vaccine development activities resulting in a set of therapeutic or prophylactic vaccines against different types of solid cancers.


Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Neoplasms/prevention & control , Neoplasms/therapy , Neovascularization, Pathologic/prevention & control , Protein Footprinting/methods , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Cancer Vaccines/biosynthesis , Cytotoxicity, Immunologic/drug effects , Endothelial Cells/chemistry , Endothelial Cells/immunology , Endothelial Cells/pathology , Hep G2 Cells , Humans , MCF-7 Cells , Neoplasms/blood supply , Neoplasms/immunology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Patents as Topic , Proteomics/methods
17.
Virus Res ; 231: 166-175, 2017 03 02.
Article En | MEDLINE | ID: mdl-27889616

Infections with sexually transmitted high-risk Human Papillomavirus (hrHPV), of which there are at least 15 genotypes, are responsible for a tremendous disease burden by causing cervical, and subsets of other ano-genital and oro-pharyngeal carcinomas, together representing 5% of all cancer cases worldwide. HPV subunit vaccines consisting of virus-like particles (VLP) self-assembled from major capsid protein L1 plus adjuvant have been licensed. Prophylactic vaccinations with the 2-valent (HPV16/18), 4-valent (HPV6/11/16/18), or 9-valent (HPV6/11/16/18/31/33/45/52/58) vaccine induce high-titer neutralizing antibodies restricted to the vaccine types that cause up to 90% of cervical carcinomas, a subset of other ano-genital and oro-pharyngeal cancers and 90% of benign ano-genital warts (condylomata). The complexity of manufacturing multivalent L1-VLP vaccines limits the number of included VLP types and thus the vaccines' spectrum of protection, leaving a panel of oncogenic mucosal HPV unaddressed. In addition, current vaccines do not protect against cutaneous HPV types causing benign skin warts, or against beta-papillomavirus (betaPV) types implicated in the development of non-melanoma skin cancer (NMSC) in immunosuppressed patients. In contrast with L1-VLP, the minor capsid protein L2 contains type-common epitopes that induce low-titer yet broadly cross-neutralizing antibodies to heterologous PV types and provide cross-protection in animal challenge models. Efforts to increase the low immunogenicity of L2 (poly)-peptides and thereby to develop broader-spectrum HPV vaccines are the focus of this review.


Antibodies, Viral/biosynthesis , Capsid Proteins/immunology , Head and Neck Neoplasms/prevention & control , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , Uterine Cervical Neoplasms/prevention & control , Vaccination , Animals , Antibodies, Neutralizing/biosynthesis , Cancer Vaccines/administration & dosage , Cancer Vaccines/biosynthesis , Cancer Vaccines/immunology , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cross Protection , Epitopes/chemistry , Epitopes/immunology , Female , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/virology , Humans , Immunity, Humoral/drug effects , Immunogenicity, Vaccine , Mice , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/genetics , Papillomaviridae/drug effects , Papillomaviridae/growth & development , Papillomaviridae/immunology , Papillomaviridae/pathogenicity , Papillomavirus Infections/immunology , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/immunology , Vaccines, Virus-Like Particle
19.
PLoS One ; 11(3): e0150623, 2016.
Article En | MEDLINE | ID: mdl-26937656

Immunotherapy approaches using checkpoint blockade, alone, or in combination with tumor antigen vaccination, or adoptive cell transfer, are emerging as promising approaches for the treatment of non-small cell lung cancer (NSCLC). In preparation for upcoming combined immunotherapy approaches in NSCLC, here, we have assessed spontaneous immune responses to XAGE-1b, a tumor specific antigen of the Cancer Testis Antigen group that has been previously reported to be spontaneously immunogenic in the Japanese population, in a cohort of Caucasian patients with NSCLC. We found spontaneous serological responses to XAGE-1b in 9% of the patients. Importantly, these responses were limited to, and represented 13% of, patients with adenocarcinoma tumors, the most frequent histological subtype, for which immunotherapy approaches are under development. Using a set of overlapping peptides spanning the entire XAGE-1b protein, and in support of the serological data, we detected significant XAGE-1b specific CD4+ T cell responses in all XAGE-1b seropositive patients and identified several CD4+ T cell epitopes. Altogether, our results support the relevance of the XAGE-1b antigen in Caucasians NSCLC patients with adenocarcinoma, and the implementation of future immunotherapies exploiting the high immunogenicity of the antigen in this patient population.


Adenocarcinoma/therapy , Antigens, Neoplasm/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Amino Acid Sequence , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/administration & dosage , Cancer Vaccines/biosynthesis , Cancer Vaccines/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Case-Control Studies , Epitopes/chemistry , Epitopes/immunology , Gene Expression , Humans , Immunotherapy, Active/methods , Interferon-gamma/agonists , Interferon-gamma/biosynthesis , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Molecular Sequence Data , Peptides/administration & dosage , Peptides/chemistry , Peptides/immunology , Primary Cell Culture
20.
Monoclon Antib Immunodiagn Immunother ; 34(6): 381-5, 2015 Dec.
Article En | MEDLINE | ID: mdl-26683176

The existence of a developed network of suppressory factors and cells against an immune response in different cancers has been proven; regulatory T cells are a typical issue. Therefore their depletion, elimination, or suppression has been assessed in different research studies that were not entirely successful. By applying an improved vaccine against regulatory T cells, we have evaluated the B cell response elicited by the vaccine in an experimental design. A previously described DNA vaccine and recombinant protein of Foxp3-Fc fusion were produced and used in the vaccination regimen. DNA construct and respective protein were injected into C57BL/6 mice. After 2 weeks, serum levels of IgG antibody and its subtypes against Foxp3 were investigated by ELISA. To produce recombinant Foxp3 for ELISA antigen coating, pET24a-Foxp3 vector was transformed into Escherichia coli strain BL21 as host cells. Afterward, protein was expressed and then purified using Ni-NTA agarose. SDS-PAGE and Western blot analysis were carried out to confirm protein expression. The expression analysis of Foxp3 was confirmed by SDS-PAGE followed by Western blot analysis. FOXP3-Fc DNA vaccine/fusion protein vaccination regimen could induce T helper-dependent humoral responses. Due to the effectiveness of Foxp3-Fc(IgG) in inducing humoral responses, it would be expected to be useful in developing vaccines in tumor therapies for the removal of regulatory T cells as a strategy for increasing the efficiency of other means of immunotherapy.


Cancer Vaccines/administration & dosage , Forkhead Transcription Factors/administration & dosage , Immunity, Humoral/drug effects , Immunoglobulin Fc Fragments/administration & dosage , Recombinant Fusion Proteins/administration & dosage , T-Lymphocytes, Regulatory/drug effects , Vaccines, DNA/administration & dosage , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cancer Vaccines/biosynthesis , Cancer Vaccines/immunology , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/biosynthesis , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Plasmids/administration & dosage , Plasmids/chemistry , Plasmids/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Vaccination , Vaccines, DNA/biosynthesis , Vaccines, DNA/immunology
...