Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 221
1.
BMC Biol ; 22(1): 88, 2024 Apr 19.
Article En | MEDLINE | ID: mdl-38641823

BACKGROUND: Immunosuppressive status is prevalent in cancer patients and increases the complexity of tumor immunotherapy. It has been found that Listeria-vectored tumor vaccines had the potential ability of two-side regulatory effect on the immune response during immunotherapy. RESULTS: The results show that the combined immunotherapy with the LM∆E6E7 and LI∆E6E7, the two cervical cancer vaccine candidate strains constructed by our lab, improves the antitumor immune response and inhibits the suppressive immune response in tumor-bearing mice in vivo, confirming the two-sided regulatory ability of the immune response caused by Listeria-vectored tumor vaccines. The immunotherapy reduces the expression level of myeloid-derived suppressor cells (MDSCs)-inducing factors and then inhibits the phosphorylation level of STAT3 protein, the regulatory factor of MDSCs differentiation, to reduce the MDSCs formation ability. Moreover, vaccines reduce the expression of functional molecules associated with MDSCs may by inhibiting the phosphorylation level of the JAK1-STAT1 and JAK2-STAT3 pathways in tumor tissues to attenuate the immunosuppressive function of MDSCs. CONCLUSIONS: Immunotherapy with Listeria-vectored cervical cancer vaccines significantly reduces the level and function of MDSCs in vivo, which is the key point to the destruction of immunosuppression. The study for the first to elucidate the mechanism of breaking the immunosuppression.


Cancer Vaccines , Myeloid-Derived Suppressor Cells , Uterine Cervical Neoplasms , Female , Humans , Mice , Animals , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Cancer Vaccines/metabolism , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/metabolism , Phosphorylation , Signal Transduction
2.
Nat Commun ; 14(1): 8047, 2023 Dec 05.
Article En | MEDLINE | ID: mdl-38052869

As key mediators of cellular communication, extracellular vesicles (EVs) have been actively explored for diagnostic and therapeutic applications. However, effective methods to functionalize EVs and modulate the interaction between EVs and recipient cells are still lacking. Here we report a facile and universal metabolic tagging technology that can install unique chemical tags (e.g., azido groups) onto EVs. The surface chemical tags enable conjugation of molecules via efficient click chemistry, for the tracking and targeted modulation of EVs. In the context of tumor EV vaccines, we show that the conjugation of toll-like receptor 9 agonists onto EVs enables timely activation of dendritic cells and generation of superior antitumor CD8+ T cell response. These lead to 80% tumor-free survival against E.G7 lymphoma and 33% tumor-free survival against B16F10 melanoma. Our study yields a universal technology to generate chemically tagged EVs from parent cells, modulate EV-cell interactions, and develop potent EV vaccines.


Cancer Vaccines , Extracellular Vesicles , Skin Neoplasms , Humans , Cancer Vaccines/metabolism , Extracellular Vesicles/metabolism , Cell Communication , Skin Neoplasms/metabolism
3.
Neoplasma ; 70(6): 747-760, 2023 Dec.
Article En | MEDLINE | ID: mdl-38014701

Vaccines composed of autophagosomes derived from tumor cells called DRibbles (DRiPs-containing blebs) are involved in the cross-presentation of tumor antigens, thus inducing cross-reactive T-cell responses against the tumor. Compared with traditional tumor lysate vaccines, autophagosome vaccines were found to be better sources of multiple tumor-associated antigens (TAAs) that activate antigen-specific T-cells. However, the involvement of tumor neoantigens in the immune responses of autophagosome vaccines remains unclear. The present study showed that exogenous autophagosome vaccines (DRibbles) combined with immune adjuvants (anti-OX40 antibody and ATP) can effectively activate functional T cells in vitro. Importantly, the combination of exogenous tumor-derived autophagosome vaccines and immune adjuvants was found to induce tumor regression in B16F10 and 4T1 tumor-bearing mice. The combination of autophagosome-enriched DRibbles with anti-OX40 antibody and ATP also exhibited optimal immune stimulation and antitumor efficiency in vivo. The effectiveness of exogenous DRibble vaccines was mainly due to their enhancement of tumor immunogenicity by increasing the presentation and release of tumor neoantigens. These findings suggest that this immunotherapeutic method may be effective in the treatment of cancer.


Cancer Vaccines , Neoplasms , Mice , Animals , Autophagosomes/metabolism , Cancer Vaccines/therapeutic use , Cancer Vaccines/metabolism , Neoplasms/metabolism , Antigens, Neoplasm/metabolism , Immunity , Adenosine Triphosphate/metabolism
4.
J Med Virol ; 95(2): e28554, 2023 02.
Article En | MEDLINE | ID: mdl-36738232

Lung cancer is a fatal disease with the highest worldwide morbidity and mortality rates. Despite recent advances in targeted therapy and immune checkpoint inhibitors for cancer, their efficacy remained limited. Therefore, we designed a Newcastle disease virus (NDV)-modified tumor whole-cell vaccine as a therapeutic vaccine and identified its antigen presentation level to develop effective immunotherapy. Then, we calculated the therapeutic and immune-stimulating effects of NDV-modified lung cancer cell vaccine and intratumoral NDV injection combination on tumor-bearing mice. The results showed that the immunogenic cell death (ICD) expression in NDV-modified lung cancer cell vaccine stimulates dendritic cell maturation and T cell activation in vivo and in vitro. Moreover, NDV-modified lung cancer cell vaccine combined with intratumoral NDV injection could significantly inhibit tumor growth and enhance the differentiation of Th1 cells and Inflammatory cell infiltration in vivo, leading to an excellent immunotherapeutic effect. Therefore, our results revealed that NDV-modified lung cancer cell vaccine combined with intratumoral NDV injection could promote antigen presentation and induce a strong antitumor immune response, which provided a promising combined therapy strategy for tumor immunotherapy.


Cancer Vaccines , Lung Neoplasms , Animals , Mice , Newcastle disease virus , Immunotherapy/methods , Cancer Vaccines/metabolism , Immunity
5.
Biomacromolecules ; 23(12): 5148-5163, 2022 Dec 12.
Article En | MEDLINE | ID: mdl-36394394

Immunotherapy is deemed one of the most powerful therapeutic approaches to treat cancer. However, limited response and tumor specificity are still major challenges to address. Herein, mannosylated polycations targeting mannose receptor- are developed as vectors for plasmid DNA (pDNA)-based vaccines to improve selective delivery of genetic material to antigen-presenting cells and enhance immune cell activation. Three diblock glycopolycations (M15A12, M29A25, and M58A45) and two triblock copolymers (M29A29B9 and M62A52B32) are generated by using mannose (M), agmatine (A), and butyl (B) derivatives to target CD206, complex nucleic acids, and favor the endosomal escape, respectively. All glycopolycations efficiently complex pDNA at N/P ratios <5, protecting the pDNA from degradation in a physiological milieu. M58A45 and M62A52B32 complexed with plasmid encoding for antigenic ovalbumin (pOVA) trigger the immune activation of cultured dendritic cells, which present the SIINFEKL antigenic peptide via specific major histocompatibility complex-I. Importantly, administration of M58A45/pOVA elicits SIINFEKL-specific T-cell response in C56BL/6 mice bearing the melanoma tumor model B16-OVA, well in line with a reduction in tumor growth. These results qualify mannosylation as an efficient strategy to target immune cells in cancer vaccination and emphasize the potential of these glycopolycations as effective delivery vehicles for nucleic acids.


Cancer Vaccines , Neoplasms , Nucleic Acids , Vaccines , Mice , Animals , Dendritic Cells , Ovalbumin , Antigen-Presenting Cells , Lymphocyte Activation , Antigen Presentation , T-Lymphocytes , Nucleic Acids/metabolism , Mice, Inbred C57BL , Cancer Vaccines/genetics , Cancer Vaccines/metabolism , Neoplasms/therapy , Neoplasms/metabolism
6.
Cell Commun Signal ; 20(1): 145, 2022 09 19.
Article En | MEDLINE | ID: mdl-36123730

BACKGROUND: Exosomes are progressively known as significant mediators of cell-to-cell communication. They convey active biomolecules to target cells and have vital functions in several physiological and pathological processes, and show substantial promise as novel treatment strategies for diseases. METHODS: In this review study, we studied numerous articles over the past two decades published on application of exosomes in different diseases as well as on perspective and challenges in this field. RESULTS: The main clinical application of exosomes are using them as a biomarker, cell-free therapeutic agents, drug delivery carriers, basic analysis for exosome kinetics, and cancer vaccine. Different exosomes from human or plant sources are utilized in various clinical trials. Most researchers used exosomes from the circulatory system for biomarker experiments. Mesenchymal stem cells (MSCs) and dendritic cells (DCs) are two widely held cell sources for exosome use. MSCs-derived exosomes are commonly used for inflammation treatment and drug delivery, while DCs-exosomes are used to induce inflammation response in cancer patients. However, the clinical application of exosomes faces various questions and challenges. In addition, translation of exosome-based clinical trials is required to conform to specific good manufacturing practices (GMP). In this review, we summarize exosomes in the clinical trials according to the type of application and disease. We also address the main questions and challenges regarding exosome kinetics and clinical applications. CONCLUSIONS: Exosomes are promising platforms for treatment of many diseases in clinical trials. This exciting field is developing hastily, understanding of the underlying mechanisms that direct the various observed roles of exosomes remains far from complete and needs further multidisciplinary research in working with these small vesicles. Video Abstract.


Cancer Vaccines , Exosomes , Biomarkers/metabolism , Cancer Vaccines/metabolism , Clinical Trials as Topic , Drug Delivery Systems , Exosomes/metabolism , Humans , Inflammation/metabolism
7.
Cancer Treat Res ; 183: 91-129, 2022.
Article En | MEDLINE | ID: mdl-35551657

Malignant tumors frequently exploit innate immunity to evade immune surveillance. The priming, function, and polarization of antitumor immunity fundamentally depends upon context provided by the innate immune system, particularly antigen presenting cells. Such context is determined in large part by sensing of pathogen specific and damage associated features by pathogen recognition receptors (PRRs). PRR activation induces the delivery of T cell priming cues (e.g. chemokines, co-stimulatory ligands, and cytokines) from antigen presenting cells, playing a decisive role in the cancer immunity cycle. Indeed, endogenous PRR activation within the tumor microenvironment (TME) has been shown to generate spontaneous antitumor T cell immunity, e.g., cGAS-STING mediated activation of antigen presenting cells after release of DNA from dying tumor cells. Thus, instigating intratumor PRR activation, particularly with the goal of generating Th1-promoting inflammation that stokes endogenous priming of antitumor CD8+ T cells, is a growing area of clinical investigation. This approach is analogous to in situ vaccination, ultimately providing a personalized antitumor response against relevant tumor associated antigens. Here I discuss clinical stage intratumor modalities that function via activation of PRRs. These approaches are being tested in various solid tumor contexts including melanoma, colorectal cancer, glioblastoma, head and neck squamous cell carcinoma, bladder cancer, and pancreatic cancer. Their mechanism (s) of action relative to other immunotherapy approaches (e.g., antigen-defined cancer vaccines, CAR T cells, dendritic cell vaccines, and immune checkpoint blockade), as well as their potential to complement these approaches are also discussed. Examples to be reviewed include TLR agonists, STING agonists, RIG-I agonists, and attenuated or engineered viruses and bacterium. I also review common key requirements for effective in situ immune activation, discuss differences between various strategies inclusive of mechanisms that may ultimately limit or preclude antitumor efficacy, and provide a summary of relevant clinical data.


Cancer Vaccines , Neoplasms , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/metabolism , Humans , Immunotherapy , Neoplasms/therapy , Receptors, Pattern Recognition/metabolism , Tumor Microenvironment
8.
J Healthc Eng ; 2022: 1964081, 2022.
Article En | MEDLINE | ID: mdl-35480145

Dendritic cells (DCs) are antigen-presenting cells that can activate T cells and initiate a primary immune response. Personalized DC vaccines have demonstrated a modest antitumor potential in some clinical pilot studies. However, those vaccines are difficult to manufacture and have a limited antitumor response. In this study, a lentiviral vector-programmed DC vaccine with high antitumor responses is developed. By transfecting with a lentiviral vector, the DC vaccine is loaded with MG-7 antigen (MG-7Ag). Three representative gastric cancer cell lines, such as KATO-3, MKN45, and SNU16, are used to estimate the in vitro cytotoxic effect of the MG-7Ag DC vaccine. Furthermore, we examine the in vivo antitumor efficacy of specific cytotoxic T lymphocytes (CTLs) induced by the MG-7Ag DC vaccine in patient-derived xenograft (PDX) mice models. The current data demonstrate that the MG-7Ag DC vaccine induced a potent CTL activity. Those CTLs have a significant cytotoxic effect on both KATO-3 and MKN45 with high level of MG-7 expression. In addition, MG-7Ag DC vaccine-mediated CTLs significantly inhibit the growth of tumor xenografts in nude mice. The MG-7Ag DC vaccine activate the cytotoxic effect of lymphocytes and can be employed as a vaccine in gastric cancer immunotherapy.


Cancer Vaccines , Stomach Neoplasms , Animals , Cancer Vaccines/metabolism , Dendritic Cells/metabolism , Humans , Mice , Mice, Nude , Stomach Neoplasms/metabolism , Stomach Neoplasms/therapy , T-Lymphocytes, Cytotoxic
9.
Front Immunol ; 13: 734256, 2022.
Article En | MEDLINE | ID: mdl-35250967

Dendritic cell (DC) vaccines have proven to be a valuable tool in cancer immune therapy. With several DC vaccines being currently tested in clinical trials, knowledge about their therapeutic value has been significantly increased in the past decade. Despite their established safety, it has become clear that objective clinical responses are not yet robust enough, requiring further optimization. Improvements of this advanced therapy medicinal product encompass, among others, regulating their immune stimulating capacity by in situ gene engineering, in addition to their implementation in combination therapy regimens. Previously, we have reported on a superior monocyte-derived DC preparation, including interleukin-15, pro-inflammatory cytokines and immunological danger signals in the culture process. These so-called IL-15 DCs have already proven to exhibit several favorable properties as cancer vaccine. Evolving research into mechanisms that could further modulate the immune response towards cancer, points to programmed death-1 as an important player that dampens anti-tumor immunity. Aiming at leveraging the immunogenicity of DC vaccines, we hypothesized that additional implementation of the inhibitory immune checkpoint molecules programmed death-ligand (PD-L)1 and PD-L2 in IL-15 DC vaccines would exhibit superior stimulatory potential. In this paper, we successfully implemented PD-L silencing at the monocyte stage in the 3-day IL-15 DC culture protocol resulting in substantial downregulation of both PD-L1 and PD-L2 to levels below 30%. Additionally, we validated that these DCs retain their specific characteristics, both at the level of phenotype and interferon gamma secretion. Evaluating their functional characteristics, we demonstrate that PD-L silencing does not affect the capacity to induce allogeneic proliferation. Ultimately designed to induce a durable tumor antigen-specific immune response, PD-L silenced IL-15 DCs were capable of surpassing PD-1-mediated inhibition by antigen-specific T cells. Further corroborating the superior potency of short-term IL-15 DCs, the combination of immune stimulatory components during DC differentiation and maturation with in situ checkpoint inhibition supports further clinical translation.


B7-H1 Antigen , Cancer Vaccines , Dendritic Cells , Neoplasms , Programmed Cell Death 1 Ligand 2 Protein , B7-H1 Antigen/genetics , CD8-Positive T-Lymphocytes , Cancer Vaccines/genetics , Cancer Vaccines/metabolism , Dendritic Cells/immunology , Humans , Interleukin-15/genetics , Neoplasms/pathology , Programmed Cell Death 1 Ligand 2 Protein/genetics
10.
Cancer Sci ; 113(3): 864-874, 2022 Mar.
Article En | MEDLINE | ID: mdl-34971473

NY-ESO-1 is a cancer/testis antigen expressed in various cancer types. However, the induction of NY-ESO-1-specific CTLs through vaccines is somewhat difficult. Thus, we developed a new type of artificial adjuvant vector cell (aAVC-NY-ESO-1) expressing a CD1d-NKT cell ligand complex and a tumor-associated antigen, NY-ESO-1. First, we determined the activation of invariant natural killer T (iNKT) and natural killer (NK) cell responses by aAVC-NY-ESO-1. We then showed that the NY-ESO-1-specific CTL response was successfully elicited through aAVC-NY-ESO-1 therapy. After injection of aAVC-NY-ESO-1, we found that dendritic cells (DCs) in situ expressed high levels of costimulatory molecules and produced interleukn-12 (IL-12), indicating that DCs undergo maturation in vivo. Furthermore, the NY-ESO-1 antigen from aAVC-NY-ESO-1 was delivered to the DCs in vivo, and it was presented on MHC class I molecules. The cross-presentation of the NY-ESO-1 antigen was absent in conventional DC-deficient mice, suggesting a host DC-mediated CTL response. Thus, this strategy helps generate sufficient CD8+ NY-ESO-1-specific CTLs along with iNKT and NK cell activation, resulting in a strong antitumor effect. Furthermore, we established a human DC-transferred NOD/Shi-scid/IL-2γcnull immunodeficient mouse model and showed that the NY-ESO-1 antigen from aAVC-NY-ESO-1 was cross-presented to antigen-specific CTLs through human DCs. Taken together, these data suggest that aAVC-NY-ESO-1 has potential for harnessing innate and adaptive immunity against NY-ESO-1-expressing malignancies.


Adjuvants, Immunologic/administration & dosage , Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Immunotherapy/methods , Membrane Proteins/administration & dosage , Adjuvants, Immunologic/metabolism , Animals , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cancer Vaccines/immunology , Cancer Vaccines/metabolism , Cross-Priming , HEK293 Cells , Humans , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , NIH 3T3 Cells , Natural Killer T-Cells/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology
11.
Int J Mol Sci ; 22(18)2021 Sep 08.
Article En | MEDLINE | ID: mdl-34575870

Bacteriophage-eukaryotic cell interaction provides the biological foundation of Phage Display technology, which has been widely adopted in studies involving protein-protein and protein-peptide interactions, and it provides a direct link between the proteins and the DNA encoding them. Phage display has also facilitated the development of new therapeutic agents targeting personalized cancer mutations. Proteins encoded by mutant genes in cancers can be processed and presented on the tumor cell surface by human leukocyte antigen (HLA) molecules, and such mutant peptides are called Neoantigens. Neoantigens are naturally existing tumor markers presented on the cell surface. In clinical settings, the T-cell recognition of neoantigens is the foundation of cancer immunotherapeutics. This year, we utilized phage display to successfully develop the 1st antibody-based neoantigen targeting approach for next-generation personalized cancer therapeutics. In this article, we discussed the strategies for identifying neoantigens, followed by using phage display to create personalized cancer therapeutics-a complete pipeline for personalized cancer treatment.


Antigens/chemistry , Bacteriophages/metabolism , Eukaryotic Cells/metabolism , Neoplasms/genetics , Neoplasms/therapy , Precision Medicine/trends , Antigens, Neoplasm/metabolism , Cancer Vaccines/immunology , Cancer Vaccines/metabolism , Drug Design , HLA Antigens/chemistry , Humans , Immunotherapy , Medical Oncology/trends , Mutation , Peptide Library , Peptides/metabolism , T-Lymphocytes/immunology
12.
Biotechnol Lett ; 43(9): 1933-1944, 2021 Sep.
Article En | MEDLINE | ID: mdl-34313864

OBJECTIVES: Human papillomavirus infection (HPV) is the most common viral infection which is causes of cervical, penal, vulvar, anal and, oropharyngeal cancer. E7 protein of HPV is a suitable target for induction of T cell responses and controlling HPV-related cancer. The aim of the current study was to designed and evaluated a novel fusion protein containing the different E7 proteins of the HPV 16, 18, 6 and 11, linked to the cell-penetrating peptide HIV-1 Tat 49-57, in order to improve cytotoxic immune responses in in-vitro and in-vivo. RESULTS: In this study whole sequence of HPV16,18,6,11 E7-Tat (47-57) and HPV16,18,6,11 E7 cloned into the vector and expressed in E. coli (BL21). The purified protein was confirmed by SDS page and western blotting and then injected into the C57BL/6 mice. The efficiency of the fusion protein vaccine was assessed by antibody response assay, cytokine assay (IL-4 and IFN-γ), CD + 8 cytotoxicity assay and tumor challenge experiment. Result showed that fusion proteins containing Adjuvant (IFA,CFA) could express higher titer of antibody. Also, we showed that vaccination with E7-Tat and, E7-Tat-ADJ induced high frequencies of E7-specific CD8 + T cells and CD107a expression as well as IFN-γ level and enhanced long-term survival in the therapeutic animal models. CONCLUSION: Our finding suggested that this novel fusion protein vaccine was able to induce therapeutic efficacy and immunogenicity by improving CD8 + T cell in TC-1 tumor bearing mice; so this vaccine may be appreciated for research against HPV and tumor immunotherapies.


Alphapapillomavirus/metabolism , HIV-1/genetics , Lung Neoplasms/virology , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/administration & dosage , Peptide Fragments/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , Alphapapillomavirus/genetics , Alphapapillomavirus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cancer Vaccines/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/growth & development , Female , HIV-1/metabolism , Human papillomavirus 11/genetics , Human papillomavirus 11/metabolism , Human papillomavirus 16/genetics , Human papillomavirus 16/metabolism , Human papillomavirus 18/genetics , Human papillomavirus 18/metabolism , Human papillomavirus 6/genetics , Human papillomavirus 6/metabolism , Humans , Lung Neoplasms/prevention & control , Mice , Mice, Inbred C57BL , Papillomavirus E7 Proteins/genetics , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Vaccination , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/metabolism
13.
Stem Cell Reports ; 16(6): 1468-1477, 2021 06 08.
Article En | MEDLINE | ID: mdl-33961792

Induced pluripotent stem cells (iPSCs) and cancer cells share cellular similarities and transcriptomic profiles. Here, we show that an iPSC-based cancer vaccine, comprised of autologous iPSCs and CpG, stimulated cytotoxic antitumor CD8+ T cell effector and memory responses, induced cancer-specific humoral immune responses, reduced immunosuppressive CD4+ T regulatory cells, and prevented tumor formation in 75% of pancreatic ductal adenocarcinoma (PDAC) mice. We demonstrate that shared gene expression profiles of "iPSC-cancer signature genes" and others are overexpressed in mouse and human iPSC lines, PDAC cells, and multiple human solid tumor types compared with normal tissues. These results support further studies of iPSC vaccination in PDAC in preclinical and clinical models and in other cancer types that have low mutational burdens.


CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Carcinoma, Pancreatic Ductal/immunology , Induced Pluripotent Stem Cells/immunology , Pancreatic Neoplasms/immunology , T-Lymphocytopenia, Idiopathic CD4-Positive/immunology , Animals , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/metabolism , Cancer Vaccines/therapeutic use , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/therapy , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunologic Memory , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/therapy , T-Lymphocytopenia, Idiopathic CD4-Positive/metabolism , Transcriptome , Xenograft Model Antitumor Assays
14.
Cells ; 10(1)2021 01 15.
Article En | MEDLINE | ID: mdl-33467606

Tissue-resident memory T (TRM) cells mediate potent local innate and adaptive immune responses and provide long-lasting protective immunity. TRM cells localize to many different tissues, including barrier tissues, and play a crucial role in protection against infectious and malignant disease. The formation and maintenance of TRM cells are influenced by numerous factors, including inflammation, antigen triggering, and tissue-specific cues. Emerging evidence suggests that these signals also contribute to heterogeneity within the TRM cell compartment. Here, we review the phenotypic and functional heterogeneity of CD8+ TRM cells at different tissue sites and the molecular determinants defining CD8+ TRM cell subsets. We further discuss the possibilities of targeting the unique cell surface molecules, cytokine and chemokine receptors, transcription factors, and metabolic features of TRM cells for therapeutic purposes. Their crucial role in immune protection and their location at the frontlines of the immune defense make TRM cells attractive therapeutic targets. A better understanding of the possibilities to selectively modulate TRM cell populations may thus improve vaccination and immunotherapeutic strategies employing these potent immune cells.


Immunologic Memory , T-Lymphocytes/cytology , ADP-ribosyl Cyclase 1/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CD8-Positive T-Lymphocytes/cytology , Cancer Vaccines/metabolism , Chemokines/metabolism , Cytokines/metabolism , Humans , Immunotherapy , Integrins/metabolism , Lectins, C-Type/metabolism , Lymphocyte Activation/immunology , Membrane Glycoproteins/metabolism , Mice , Neoplasms/metabolism , Phenotype , Receptors, Cytokine/metabolism , Signal Transduction , Transcription Factors
15.
Biosci Rep ; 40(12)2020 12 23.
Article En | MEDLINE | ID: mdl-33169789

Immunotherapy is a breakthrough approach for cancer treatment and prevention. By exploiting the fact that cancer cells have overexpression of tumor antigens responsible for its growth and progression, which can be identified and removed by boosting the immune system. In silico techniques have provided efficient ways for developing preventive measures to ward off cancer. Herein, we have designed a potent cytotoxic T-lymphocyte epitope to elicit a desirable immune response against carcinogenic melanoma-associated antigen-A11. Potent epitope was predicted using reliable algorithms and characterized by advanced computational avenue CABS molecular dynamics simulation, for full flexible binding with HLA-A*0201 and androgen receptor to large-scale rearrangements of the complex system. Results showed the potent immunogenic construct (KIIDLVHLL), from top epitopes using five algorithms. Molecular docking analyses showed the strong binding of epitope with HLA-A*0201 and androgen receptor with docking score of -780.6 and -641.06 kcal/mol, respectively. Molecular dynamics simulation analysis revealed strong binding of lead epitope with androgen receptor by involvement of 127 elements through atomic-model study. Full flexibility study showed stable binding of epitope with an average root mean square deviation (RMSD) 2.21 Å and maximum RMSD value of 6.48 Å in optimal cluster density area. The epitope also showed remarkable results with radius of gyration 23.0777 Å, world population coverage of 39.08% by immune epitope database, and transporter associated with antigen processing (TAP) affinity IC50 value of 2039.65 nm. Moreover, in silico cloning approach confirmed the expression and translation capacity of the construct within a suitable expression vector. The present study paves way for a potential immunogenic construct for prevention of cancer.


Antigens, Neoplasm/therapeutic use , Cancer Vaccines/therapeutic use , Cytotoxicity, Immunologic , Drug Design , Epitopes, T-Lymphocyte , Neoplasm Proteins/therapeutic use , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Algorithms , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/metabolism , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , Immunogenicity, Vaccine , Molecular Docking Simulation , Molecular Dynamics Simulation , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Receptors, Androgen/immunology , Receptors, Androgen/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Subunit/metabolism , Vaccines, Subunit/therapeutic use
16.
J Immunother ; 43(9): 273-282, 2020.
Article En | MEDLINE | ID: mdl-32925563

Expression of carbonic-anhydrase IX (CAIX) in clear cell renal cell carcinoma (RCC) makes it an attractive vaccine target. We developed a fusion-gene construct, granulocyte-macrophage (GM) colony-stimulating factor+CAIX, delivered by an adenoviral vector (Ad) into autologous dendritic cells (DCs) in this phase 1 study. The injected immature DCs were expected to stimulate an antigen-specific immune response against CAIX expressing RCC. Three dose-escalation cohorts (5, 15, and 50×10 cells/administration) were injected intradermally q2wk×3 doses based on a 3+3 design. The primary objective was the safety of the injections. Secondary objectives were immune responses using enzyme-linked immunosorbent spot, a serum biomarker panel, and clinical response. Fifteen patients with metastatic RCC were enrolled, and 9 patients received all 3 doses. No serious adverse events were seen. There were 3 (33%) patients with grade 1 fatigue, 1 of whom subsequently experienced grade 2 fatigue. One patient (11%) experienced grade 1-2 leukopenia. Only 1 patient (11%) experienced grade 2 flu-like symptoms. Of the 9 patients who received treatment, 1 expired of progressive disease, 2 patients were lost to follow-up and 6 patients are alive. Of the 6 patients, 5 have progressive disease, and 1 has completed treatment with stable disease at 27 months follow-up. Immune response measurements appeared more robust in higher dose cohorts, which appeared to be related to patients with stable disease at 3 months. These early data show that autologous immature DC-AdGMCAIX can be safely given to metastatic RCC patients without any serious adverse events with CAIX-specific immune response elicited by the treatment. These preliminary data support further study of Ad-GMCAIX, particularly with combination therapies that may enhance clinical activity.


Antigens, Neoplasm/genetics , Cancer Vaccines/administration & dosage , Carbonic Anhydrase IX/genetics , Carcinoma, Renal Cell/therapy , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Kidney Neoplasms/therapy , Antigens, Neoplasm/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/genetics , Cancer Vaccines/metabolism , Carbonic Anhydrase IX/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Dendritic Cells/metabolism , Disease Management , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Treatment Outcome
17.
Eur Rev Med Pharmacol Sci ; 24(13): 7454-7461, 2020 07.
Article En | MEDLINE | ID: mdl-32706085

OBJECTIVE: To develop a promising approach for tumor immunotherapy with G250 antigen-based DNA vaccine and to investigate its anti-tumor response in mice with renal cell carcinoma. MATERIALS AND METHODS: G250 derived from human, monkey and mouse were prepared by PCR. The heterogeneous chimeric G250 gene was obtained by integrating different gene fragments of three species. Then, the chimeric G250 was inserted into a eukaryotic expression plasmid pVAX1-IRES-GM/B7 to obtain DNA vaccine (named pVAX1-tG250-GM/B7) which could express chimeric G250 antigen and immune adjuvants simultaneously. By transfecting into Cos7 cells, the expression of chimeric G250 antigen was tested using flow cytometry and immunofluorescence assay. The immunological response and protection against tumor were evaluated in vivo. RESULTS: Recombinant plasmid DNA vaccine was constructed successfully through identification of PCR and gene sequencing. The chimeric G250 antigen was well expressed in Cos7 cells. A strong immune response can be detected through ELISPOT and ELISA induced by pVAX1-tG250-GM/B7. The mice vaccinated with pVAX1-tG250-GM/B7, balb/c showed significant inhibition of tumor and a longer time of survival compared with control group. CONCLUSIONS: The experimental results of this study exhibited that the DNA vaccine based on heterogeneous chimeric antigen can produce efficient anti-tumor effect in vivo and they represent a promising strategy for tumor immunotherapy.


Antigens, Neoplasm/pharmacology , Cancer Vaccines/pharmacology , Carbonic Anhydrase IX/pharmacology , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , COS Cells , Cancer Vaccines/genetics , Cancer Vaccines/metabolism , Carbonic Anhydrase IX/genetics , Carbonic Anhydrase IX/metabolism , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Chlorocebus aethiops , Cytokines/metabolism , Female , Haplorhini , Immunogenicity, Vaccine , Immunoglobulin G/blood , Kidney Neoplasms/immunology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , Tumor Burden/drug effects , Vaccination , Vaccines, DNA/genetics , Vaccines, DNA/metabolism , Vaccines, DNA/pharmacology
18.
Nat Commun ; 11(1): 1110, 2020 02 28.
Article En | MEDLINE | ID: mdl-32111828

Targeted delivery of a nanovaccine loaded with a tumor antigen and adjuvant to the lymph nodes (LNs) is an attractive approach for improving cancer immunotherapy outcomes. However, the application of this technique is restricted by the paucity of suitable tumor-associated antigens (TAAs) and the sophisticated technology required to identify tumor neoantigens. Here, we demonstrate that a self-assembling melittin-lipid nanoparticle (α-melittin-NP) that is not loaded with extra tumor antigens promotes whole tumor antigen release in situ and results in the activation of antigen-presenting cells (APCs) in LNs. Compared with free melittin, α-melittin-NPs markedly enhance LN accumulation and activation of APCs, leading to a 3.6-fold increase in antigen-specific CD8+ T cell responses. Furthermore, in a bilateral flank B16F10 tumor model, primary and distant tumor growth are significantly inhibited by α-melittin-NPs, with an inhibition rate of 95% and 92%, respectively. Thus, α-melittin-NPs induce a systemic anti-tumor response serving as an effective LN-targeted whole-cell nanovaccine.


Cancer Vaccines/immunology , Drug Delivery Systems , Lymph Nodes/immunology , Melitten/administration & dosage , Nanoparticles/administration & dosage , Neoplasms/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemistry , Cancer Vaccines/metabolism , Cell Line, Tumor , Cytokines/immunology , Female , Immunotherapy , Lipids/administration & dosage , Lipids/chemistry , Lymph Nodes/metabolism , Melitten/chemistry , Melitten/immunology , Melitten/metabolism , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Nanoparticles/metabolism , Neoplasms/therapy , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
19.
Nat Commun ; 11(1): 1137, 2020 02 28.
Article En | MEDLINE | ID: mdl-32111835

The interaction between immune cells and phosphatidylserine (PS) molecules exposed on the surface of apoptotic-tumor bodies, such as those induced by chemotherapies, contributes to the formation of an immunosuppressive tumor microenvironment (TME). Annexin A5 (AnxA5) binds with high affinity to PS externalized by apoptotic cells, thereby hindering their interaction with immune cells. Here, we show that AnxA5 administration rescue the immunosuppressive state of the TME induced by chemotherapy. Due to the preferential homing of AnxA5 to the TME enriched with PS+ tumor cells, we demonstrate in vivo that fusing tumor-antigen peptide to AnxA5 significantly enhances its immunogenicity and antitumor efficacy when administered after chemotherapy. Also, the therapeutic antitumor effect of an AnxA5-peptide fusion can be further enhanced by administration of other immune checkpoint inhibitors. Our findings support the administration of AnxA5 following chemotherapy as a promising immune checkpoint inhibitor for cancer treatment.


Annexin A5/therapeutic use , Cancer Vaccines/therapeutic use , Immunologic Factors/therapeutic use , Neoplasms/therapy , Animals , Annexin A5/genetics , Annexin A5/metabolism , Antibodies, Blocking/therapeutic use , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/therapeutic use , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/metabolism , Cell Line, Tumor , Cisplatin/adverse effects , Cisplatin/therapeutic use , Disease Models, Animal , Female , Humans , Immunologic Factors/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/immunology , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Papillomavirus E7 Proteins/therapeutic use , Phosphatidylserines/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
20.
Sci Rep ; 10(1): 3764, 2020 02 28.
Article En | MEDLINE | ID: mdl-32111878

A critical limitation of Salmonella typhimurium (S. typhimurium) as an anti-cancer agent is the loss of their invasive or replicative activities, which results in no or less delivery of anti-cancer agents inside cancer cells in cancer therapy. Here we developed an oxytolerant attenuated Salmonella strain (KST0650) from the parental KST0649 (ΔptsIΔcrr) strain using radiation mutation technology (RMT). The oxytolerant KST0650 strain possessed 20-times higher replication activity in CT26 cancer cells and was less virulent than KST0649. Furthermore, KST0650 migrated effectively into tumor tissues in mice. KST0650 was further equipped with a plasmid harboring a spliced form of the intracellular pro-apoptotic protein sATF6, and the expression of sATF6 was controlled by the radiation-inducible recN promoter. The new strain was named as KST0652, in which sATF6 protein expression was induced in response to radiation in a dose-dependent manner. This strain was effectively delivered inside cancer cells and tumor tissues via the Salmonella type III secretion system (T3SS). In addition, combination treatment with KST0652 and radiation showed a synergistic anti-tumor effect in murine tumor model with complete inhibition of tumor growth and protection against death. In conclusion, we showed that RMT can be used to effectively develop an anti-tumor Salmonella strain for delivering anti-cancer agents inside tumors.


Activating Transcription Factor 6 , Cancer Vaccines , Mutation , Neoplasm Proteins , Neoplasms, Experimental , Salmonella typhimurium , Type III Secretion Systems , Activating Transcription Factor 6/biosynthesis , Activating Transcription Factor 6/genetics , Animals , Cancer Vaccines/genetics , Cancer Vaccines/metabolism , Cell Line, Tumor , Male , Mice , Mice, Inbred BALB C , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/microbiology , Neoplasms, Experimental/therapy , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism
...