Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 776
Filter
1.
Drug Dev Res ; 85(5): e22244, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39138855

ABSTRACT

Nanovaccines have been designed to overcome the limitations associated with conventional vaccines. Effective delivery methods such as engineered carriers or smart nanoparticles (NPs) are critical requisites for inducing self-tolerance and optimizing vaccine immunogenicity with minimum side effects. NPs can be used as adjuvants, immunogens, or nanocarriers to develop nanovaccines for efficient antigen delivery. Multiloaded nanovaccines carrying multiple tumor antigens along with immunostimulants can effectively increase immunity against tumor cells. They can be biologically engineered to boost interactions with dendritic cells and to allow a gradual and constant antigen release. Modifying NPs surface properties, using high-density lipoprotein-mimicking nanodiscs, and developing nano-based artificial antigen-presenting cells such as dendritic cell-derived-exosomes are amongst the new developed technologies to enhance antigen-presentation and immune reactions against tumor cells. The present review provides an overview on the different perspectives, improvements, and barriers of successful clinical application of current cancer therapeutic and vaccination options. The immunomodulatory effects of different types of nanovaccines and the nanoparticles incorporated into their structure are described. The advantages of using nanovaccines to prevent and treat common illnesses such as AIDS, malaria, cancer and tuberculosis are discussed. Further, potential paths to develop optimal cancer vaccines are described. Given the immunosuppressive characteristics of both cancer cells and the tumor microenvironment, applying immunomodulators and immune checkpoint inhibitors in combination with other conventional anticancer therapies are necessary to boost the effectiveness of the immune response.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Immunotherapy , Nanoparticles , Neoplasms , Humans , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , Nanoparticles/administration & dosage , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/administration & dosage , Animals , Combined Modality Therapy , Drug Delivery Systems/methods , Nanovaccines
2.
J Control Release ; 373: 358-369, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39009083

ABSTRACT

Cancer vaccines based on single-source (exogenous or endogenous) tumor-associated antigens (TAAs) are often challenged by the insufficient T cell response and the immunosuppressive tumor microenvironment (TME). Herein, a dual TAAs-boosted nanovaccine based on cancer cell (4T1) membrane-cloaked, CO-immobilized Prussian blue nanoparticles (4T1-PB-CO NPs) is developed and coupled with anti-interleukin (IL)-10 therapy to maximize the efficacy of antitumor immunotherapy. 4T1 cell membrane not only endows NPs with tumor targeting ability, but also serves as exogenous TAAs to trigger CD4+ T cell response and M1-phenotype polarization of tumor-associated macrophages. Under near-infrared light irradiation, 4T1-PB-CO NPs release CO to induce immunogenic cell death (ICD) of tumor cells, thus generating endogenous TAAs to activate CD8+ T cell response. Meanwhile, ICD triggers release of damage-associated molecular patterns, which can promote DC maturation to amplify the antitumor T cell response. When combined with anti-IL-10 that reverses the immunosuppressive TME, 4T1-PB-CO NPs efficiently suppress the primary tumors and produce an abscopal effect to inhibit distant tumors in a breast tumor-bearing mouse model. Such a two-pronged cancer vaccine represents a promising paradigm for robust antitumor immunotherapy.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Mice, Inbred BALB C , Nanoparticles , Tumor Microenvironment , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Female , Cell Line, Tumor , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Ferrocyanides/chemistry , Interleukin-10/immunology , Mice , Immunotherapy/methods , CD8-Positive T-Lymphocytes/immunology , Nanovaccines
3.
Int J Pharm ; 660: 124254, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38795934

ABSTRACT

Cancer vaccines can be utilized in combination with checkpoint inhibitors to optimally stimulate the anti-tumor immune response. Uptake of vaccine antigen by antigen presenting cells (APCs) is a prerequisite for T cell priming, but often relies on non-specific mechanisms. Here, we have developed a novel vaccination strategy consisting of cancer antigen-containing liposomes conjugated with CD169- or DC-SIGN-specific nanobodies (single domain antibodies) to achieve specific uptake by APCs. Our studies demonstrate efficient nanobody liposome uptake by human and murine CD169+ and DC-SIGN+ APCs in vitro and in vivo when compared to control liposomes or liposomes with natural ligands for CD169 and DC-SIGN. Uptake of CD169 nanobody liposomes resulted in increased T cell activation by human APCs and stimulated naive T cell priming in mouse models. In conclusion, while nanobody liposomes have previously been utilized to direct drugs to tumors, here we show that nanobody liposomes can be applied as vaccination strategy that can be extended to other receptors on APCs in order to elicit a potent immune response against tumor antigens.


Subject(s)
Antigen-Presenting Cells , Cancer Vaccines , Liposomes , Mice, Inbred C57BL , Single-Domain Antibodies , T-Lymphocytes , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Single-Domain Antibodies/immunology , Single-Domain Antibodies/administration & dosage , Humans , T-Lymphocytes/immunology , Mice , Antigen-Presenting Cells/immunology , Female , Antigens, Neoplasm/immunology , Antigens, Neoplasm/administration & dosage , Lymphocyte Activation/drug effects
4.
Biomater Sci ; 10(11): 2865-2876, 2022 May 31.
Article in English | MEDLINE | ID: mdl-35445677

ABSTRACT

Liver cancer is the most common malignant tumor and liver cancer immunotherapy has been one of the research hotspots. To induce antigen-specific antitumor immune responses against liver cancer, we developed antigen and adjuvant co-delivery nanovaccines (APPCs). Polyanionic alginate (ALG) and polycationic polyethyleneimine (PEI) were utilized to co-deliver a glypican-3 peptide antigen and an unmethylated cytosine-phosphate-guanine (CpG) adjuvant by electrostatic interactions. A cellular uptake study confirmed that APPC could promote antigen and adjuvant uptake by dendritic cells (DCs). Importantly, APPC facilitated the endosomal escape of the peptide for antigen delivery into the cytoplasm. In addition, APPC showed significant stimulation of DC maturation in vitro. APPC could also efficiently prime DCs and induce cytotoxic T lymphocyte responses in vivo. The in vitro cell viability assay and the in vivo histocompatibility showed that APPC was non-toxic within the tested concentration. This study demonstrates that the peptide antigen and the CpG adjuvant co-delivery nanovaccine have potential applications in liver cancer immunotherapy.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Liver Neoplasms , Nanoparticles , Toll-Like Receptor 9 , Adjuvants, Immunologic/administration & dosage , Alginates/administration & dosage , Animals , Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunotherapy , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Peptides/administration & dosage , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/metabolism
5.
J Immunother Cancer ; 10(4)2022 04.
Article in English | MEDLINE | ID: mdl-35428705

ABSTRACT

BACKGROUND: Type 1 conventional dendritic cells (cDC1s) are characterized by their ability to induce potent CD8+ T cell responses. In efforts to generate novel vaccination strategies, notably against cancer, human cDC1s emerge as an ideal target to deliver antigens. cDC1s uniquely express XCR1, a seven transmembrane G protein-coupled receptor. Due to its restricted expression and endocytic nature, XCR1 represents an attractive receptor to mediate antigen-delivery to human cDC1s. METHODS: To explore tumor antigen delivery to human cDC1s, we used an engineered version of XCR1-binding lymphotactin (XCL1), XCL1(CC3). Site-specific sortase-mediated transpeptidation was performed to conjugate XCL1(CC3) to an analog of the HLA-A*02:01 epitope of the cancer testis antigen New York Esophageal Squamous Cell Carcinoma-1 (NY-ESO-1). While poor epitope solubility prevented isolation of stable XCL1-antigen conjugates, incorporation of a single polyethylene glycol (PEG) chain upstream of the epitope-containing peptide enabled generation of soluble XCL1(CC3)-antigen fusion constructs. Binding and chemotactic characteristics of the XCL1-antigen conjugate, as well as its ability to induce antigen-specific CD8+ T cell activation by cDC1s, was assessed. RESULTS: PEGylated XCL1(CC3)-antigen conjugates retained binding to XCR1, and induced cDC1 chemoattraction in vitro. The model epitope was efficiently cross-presented by human cDC1s to activate NY-ESO-1-specific CD8+ T cells. Importantly, vaccine activity was increased by targeting XCR1 at the surface of cDC1s. CONCLUSION: Our results present a novel strategy for the generation of targeted vaccines fused to insoluble antigens. Moreover, our data emphasize the potential of targeting XCR1 at the surface of primary human cDC1s to induce potent CD8+ T cell responses.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Dendritic Cells , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Lymphokines , Membrane Proteins , Sialoglycoproteins , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cross-Priming , Dendritic Cells/immunology , Epitopes/immunology , Esophageal Neoplasms/immunology , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/therapy , Humans , Lymphokines/administration & dosage , Lymphokines/immunology , Male , Membrane Proteins/administration & dosage , Membrane Proteins/immunology , Sialoglycoproteins/administration & dosage , Sialoglycoproteins/immunology
6.
Cancer Sci ; 113(3): 864-874, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34971473

ABSTRACT

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.


Subject(s)
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
7.
Sci Rep ; 11(1): 19794, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34611284

ABSTRACT

Adoptive T-cell transfer (ACT) offers a curative therapeutic option for subsets of melanoma and hematological cancer patients. To increase response rates and broaden the applicability of ACT, it is necessary to improve the post-infusion performance of the transferred T cells. The design of improved treatment strategies includes transfer of cells with a less differentiated phenotype. Such T cell subsets have high proliferative potential but require stimulatory signals in vivo to differentiate into tumor-reactive effector T cells. Thus, combination strategies are needed to support the therapeutic implementation of less differentiated T cells. Here we show that systemic delivery of tumor-associated antigens (TAAs) facilitates in vivo priming and expansion of previously non-activated T cells and enhance the cytotoxicity of activated T cells. To achieve this in vivo priming, we use flexible delivery vehicles of TAAs and a TLR7/8 agonist. Contrasting subcutaneous delivery systems, these vehicles accumulate TAAs in the spleen, thereby achieving close proximity to both cross-presenting dendritic cells and transferred T cells, resulting in robust T-cell expansion and anti-tumor reactivity. This TAA delivery platform offers a strategy to safely potentiate the post-infusion performance of T cells using low doses of antigen and TLR7/8 agonist, and thereby enhance the effect of ACT.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunotherapy, Adoptive , Neoplasms/immunology , Neoplasms/therapy , Animals , Antigen Presentation , Antigens, Neoplasm/administration & dosage , Biomarkers , Combined Modality Therapy , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Drug Delivery Systems , Epitopes/administration & dosage , Epitopes/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunomodulation , Immunotherapy, Adoptive/methods , Interferon Type I/biosynthesis , Liposomes , Lymphocyte Activation/immunology , Neoplasms/diagnosis , Neoplasms/mortality , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome , Tumor Escape/immunology
8.
Nat Commun ; 12(1): 5195, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34465781

ABSTRACT

Functional tumor-specific cytotoxic T cells elicited by therapeutic cancer vaccination in combination with oncolytic viruses offer opportunities to address resistance to checkpoint blockade therapy. Two cancer vaccines, the self-adjuvanting protein vaccine KISIMA, and the recombinant oncolytic vesicular stomatitis virus pseudotyped with LCMV-GP expressing tumor-associated antigens, termed VSV-GP-TAA, both show promise as a single agent. Here we find that, when given in a heterologous prime-boost regimen with an optimized schedule and route of administration, combining KISIMA and VSV-GP-TAA vaccinations induces better cancer immunity than individually. Using several mouse tumor models with varying degrees of susceptibility for viral replication, we find that priming with KISIMA-TAA followed by VSV-GP-TAA boost causes profound changes in the tumor microenvironment, and induces a large pool of poly-functional and persistent antigen-specific cytotoxic T cells in the periphery. Combining this heterologous vaccination with checkpoint blockade further improves therapeutic efficacy with long-term survival in the spectrum. Overall, heterologous vaccination with KISIMA and VSV-GP-TAA could sensitize non-inflamed tumors to checkpoint blockade therapy.


Subject(s)
Cancer Vaccines/immunology , Neoplasms/immunology , Neoplasms/therapy , Oncolytic Viruses/immunology , Vesicular stomatitis Indiana virus/immunology , Animals , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Combined Modality Therapy , Female , Humans , Mice , Mice, Inbred C57BL , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment , Vaccination , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/physiology , Virus Replication
9.
Mol Pharm ; 18(10): 3832-3842, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34499836

ABSTRACT

Use of tumor-associated antigens for cancer immunotherapy is limited due to their poor in vivo stability and low cellular uptake. Delivery of antigenic peptides using synthetic polymer-based nanostructures has been actively pursued but with limited success. Peptide-based nanostructures hold much promise as delivery vehicles due to their easy design and synthesis and inherent biocompatibility. Here, we report self-assembly of a dipeptide containing a non-natural amino acid, α,ß-dehydrophenylalanine (ΔF), into nanotubes, which efficiently entrapped a MAGE-3-derived peptide (M3). M3 entrapped in F-ΔF nanotubes was more stable to a nonspecific protease treatment and both F-ΔF and F-ΔF-M3 showed no cellular toxicity for four cancerous and noncancerous cell lines used. F-ΔF-M3 showed significantly higher cellular uptake in RAW 267.4 macrophage cells compared to M3 alone and also induced in vitro maturation of dendritic cells (DCs). Immunization of mice with F-ΔF-M3 selected a higher number of IFN-γ secreting CD8+ T cells and CD4+ T compared to M3 alone. On day 21, a tumor growth inhibition ratio (TGI, %) of 41% was observed in a murine melanoma model. These results indicate that F-ΔF nanotubes are highly biocompatible, efficiently delivered M3 to generate cytotoxic T lymphocytes responses, and able to protect M3 from degradation under in vivo conditions. The F-ΔF dipeptide-based nanotubes may be considered as a good platform for further development as delivery agents.


Subject(s)
Antigens, Neoplasm/administration & dosage , Nanoparticle Drug Delivery System/administration & dosage , Testis/immunology , Animals , Humans , Immunotherapy/methods , MCF-7 Cells , Male , Melanoma/immunology , Melanoma/therapy , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Nanotubes, Peptide , Neoplasm Transplantation , RAW 264.7 Cells
10.
Immunol Lett ; 239: 77-87, 2021 11.
Article in English | MEDLINE | ID: mdl-34508790

ABSTRACT

Cancer immunotherapy is emerging as a viable treatment option for several types of cancer. Active immunotherapy aims for the induction of specific antitumor immune responses; this goal requires strategies capable of increasing the immunogenicity of tumour antigens. Parvovirus B19 virus-like particles (B19-VLPs) formed of VP2 protein had been shown to be an effective multi-neoepitope delivery system capable of inducing specific cellular responses towards coupled antigens and reducing tumour growth and lung metastases in triple negative breast cancer mouse model. These findings encouraged us to further characterise these VP2 B19-VLPs by testing their capacity to simultaneously induce cellular and humoral responses towards other tumour-associated antigens, as this had not yet been evaluated. Here, we designed and evaluated in the 4T1 breast cancer model the prophylactic and therapeutic effect of VP2 B19-VLPs decorated with cellular (P53) and humoral (MUC1) epitopes. Balb/c mice were immunised with chimaeric VLPs, vehicle, or VLPs plus adjuvant. Tumour establishment and growth, lung metastasis, and cellular and humoral immune responses were evaluated. The prophylactic administration of chimaeric VLPs without adjuvant prevented the establishment of the tumour, while by therapeutic administration, chimaeric VLPs induced smaller tumour growth and decreased the number of metastases in the lung compared to wild-type VLPs. chimaeric VLPs induced high antibody titres towards the MUC1 epitope, as well as specific cellular responses towards P53 epitopes in lymph nodes local to the tumour. Our results reinforce and extend the utility of VP2 B19-VLPs as an encouraging tumour antigen delivery system in cancer immunotherapy able to improve tumour immunity in TNBC by inducing cellular and humoral immune responses.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Parvovirus B19, Human/immunology , Triple Negative Breast Neoplasms/therapy , Vaccines, Virus-Like Particle/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Neoplasm/administration & dosage , Bacillus thuringiensis Toxins/administration & dosage , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Disease Models, Animal , Endotoxins/administration & dosage , Female , Hemolysin Proteins/administration & dosage , Humans , Immunity, Cellular , Immunity, Humoral , Immunogenicity, Vaccine , Insect Proteins , Mice , Receptors, Cell Surface , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Vaccines, Virus-Like Particle/administration & dosage
11.
J Clin Invest ; 131(16)2021 08 16.
Article in English | MEDLINE | ID: mdl-34396986

ABSTRACT

Therapeutic vaccines that augment T cell responses to tumor antigens have been limited by poor potency in clinical trials. In contrast, the transfer of T cells modified with foreign transgenes frequently induces potent endogenous T cell responses to epitopes in the transgene product, and these responses are undesirable, because they lead to rejection of the transferred T cells. We sought to harness gene-modified T cells as a vaccine platform and developed cancer vaccines composed of autologous T cells modified with tumor antigens and additional adjuvant signals (Tvax). T cells expressing model antigens and a broad range of tumor neoantigens induced robust and durable T cell responses through cross-presentation of antigens by host DCs. Providing Tvax with signals such as CD80, CD137L, IFN-ß, IL-12, GM-CSF, and FLT3L enhanced T cell priming. Coexpression of IL-12 and GM-CSF induced the strongest CD4+ and CD8+ T cell responses through complimentary effects on the recruitment and activation of DCs, mediated by autocrine IL-12 receptor signaling in the Tvax. Therapeutic vaccination with Tvax and adjuvants showed antitumor activity in subcutaneous and metastatic preclinical mouse models. Human T cells modified with neoantigens readily activated specific T cells derived from patients, providing a path for clinical translation of this therapeutic platform in cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Adjuvants, Immunologic/administration & dosage , Allografts , Animals , Antigen Presentation , Antigens, Neoplasm/administration & dosage , Autografts , CD8-Positive T-Lymphocytes/transplantation , Cancer Vaccines/immunology , Cross Reactions/immunology , Dendritic Cells/immunology , Female , Humans , Immunologic Memory , Immunotherapy, Adoptive , Interleukin-12/immunology , Lymphoid Tissue/immunology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Translational Research, Biomedical
12.
Immunol Lett ; 239: 32-41, 2021 11.
Article in English | MEDLINE | ID: mdl-34418488

ABSTRACT

Tumor-derived exosomes (TEXs) could be harnessed as an immunotherapeutic cancer vaccine. These nanovesicles are inherently possesses rich tumor antigen reservoirs. Due to their undesirable features such as poor or limited immunogenicity as well as facilitation of cancer development via mediating communication between tumor cells TEXs could be transformed into an effective immune adjuvant delivery system that initiates a strong humoral and cell-mediated tumor-specific immune response. Engineering TEXs to harbor immunostimulatory molecules still remains a challenge. Previously, we demonstrated that nucleic acid ligand encapsulated liposomes could trigger synergistic strong humoral, and cell mediated immune responses and provokes tumor regression to that of their standalone counterparts. In this study, we evaluated to immunogenicity of 4T1/Her2 cell-derived exosomes upon loading them with two potent immuno adjuvant, a TLR9 ligand, K-type CpG ODN and a TLR3 ligand, p(I:C). Engineered TEXs co-encapsulating both ligands displayed boosted immunostimulatory properties by activating antigen-specific primary and memory T cell responses. Furthermore, our exosome-based vaccine candidate elicited robust Th1-biased immunity as evidenced by elevated secretion of IgG2a and IFNγ. In a therapeutic cancer model, administration of4T1 tumor derived exosomes loaded with CpG ODN and p(I:C) to animals regress tumor growth in 4T1 tumor-bearing mice. Taken together this work implicated that an exosome-based therapeutic vaccine promoted strong cellular and humoral anti-tumor immunity that is sufficient to reverse established tumors. This approach offers a personalized tumor therapy strategy that could be implemented in the clinic.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Neoplasm/administration & dosage , Breast Neoplasms/therapy , Cancer Vaccines/administration & dosage , Exosomes/immunology , Animals , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cancer Vaccines/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Humans , Memory T Cells/immunology , Mice , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Poly I-C/administration & dosage , Poly I-C/immunology , Th1 Cells/immunology , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 9/metabolism
13.
Front Immunol ; 12: 705974, 2021.
Article in English | MEDLINE | ID: mdl-34305947

ABSTRACT

Antigen-specific immunotherapies, in particular peptide vaccines, depend on the recognition of naturally presented antigens derived from mutated and unmutated gene products on human leukocyte antigens, and represent a promising low-side-effect concept for cancer treatment. So far, the broad application of peptide vaccines in cancer patients is hampered by challenges of time- and cost-intensive personalized vaccine design, and the lack of neoepitopes from tumor-specific mutations, especially in low-mutational burden malignancies. In this study, we developed an immunopeptidome-guided workflow for the design of tumor-associated off-the-shelf peptide warehouses for broadly applicable personalized therapeutics. Comparative mass spectrometry-based immunopeptidome analyses of primary chronic lymphocytic leukemia (CLL) samples, as representative example of low-mutational burden tumor entities, and a dataset of benign tissue samples enabled the identification of high-frequent non-mutated CLL-associated antigens. These antigens were further shown to be recognized by pre-existing and de novo induced T cells in CLL patients and healthy volunteers, and were evaluated as pre-manufactured warehouse for the construction of personalized multi-peptide vaccines in a first clinical trial for CLL (NCT04688385). This workflow for the design of peptide warehouses is easily transferable to other tumor entities and can provide the foundation for the development of broad personalized T cell-based immunotherapy approaches.


Subject(s)
Antigens, Neoplasm , Epitopes , Immunotherapy , Leukemia, Lymphocytic, Chronic, B-Cell , Peptides , Adult , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , Epitopes/administration & dosage , Epitopes/immunology , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Male , Peptides/administration & dosage , Peptides/immunology
14.
Anticancer Res ; 41(8): 4101-4115, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34281881

ABSTRACT

BACKGROUND/AIM: Neoantigens are tumor-specific antigens that emerge due to gene mutations in tumor cells, and are highly antigenic epitopes that escape central immune tolerance in the thymus, making cancer vaccine therapy a desirable option. PATIENTS AND METHODS: Tumor neoantigens were predicted in 17 patients with advanced cancer. They were resistant to the standard treatment regime, and their synthetic peptides were pulsed to the patient's monocyte-derived dendritic cells (DCs), and administered to the patient's lymph nodes via ultrasound. RESULTS: Some patients showed sustained tumor shrinkage after this treatment, while some did not respond, showing no ELISpot reaction. Although the number of mutations and the predicted neoantigen epitopes differed between patients, the clinical effect depended more on the presence or absence of an immune response after vaccination rather than the number of neoantigens. CONCLUSION: Intranodal neoantigen peptide-pulsed DC vaccine administration therapy has clinical and immunological efficacy and safety.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/therapeutic use , Dendritic Cells , Neoplasms/therapy , Peptides/administration & dosage , Adult , Aged , Female , Humans , Immunotherapy , Male , Middle Aged , Retrospective Studies , Treatment Outcome
15.
Sci Rep ; 11(1): 14661, 2021 07 19.
Article in English | MEDLINE | ID: mdl-34282215

ABSTRACT

Lack of pre-existing tumor infiltrated T cells resulting in resistance to programmed cell death protein 1 (PD-1) blockade therapies can be solved by combining with anti-cancer vaccines and CpG-ODN in increasing T cell expansion and infiltration. Therefore, we prepared an ex vivo dendritic cell-based (DC) vaccine pulsed with a low dose of either liposomal or non-liposomal gp100 antigen (2.8 µg) plus CpG-ODN (800 ng) formulations and evaluated its anti-tumor activity in combination with anti-PD-1 therapy. Our results showed a combination of liposomal peptide plus CpG-ODN pulsed DC with anti-PD-1 antibody was more efficacious, as evidenced by a significant increase in Teff/Treg TILs with a marked fourfold elevation of IFN-γ expression level in the tumor site of treated mice which reversed resistance to PD-1 blockade in a CD8 T cell-dependent manner. Furthermore, this combination also led to a remarkable tumor remission and prolonged survival rate in melanoma-bearing mice compared to non-liposomal peptide plus CpG-ODN or single-treated liposomal peptide formulations. Our results provide essential insights to devise combining regimens to improve the efficacy of immune checkpoint blockers even by a low dose of peptide and CpG-ODN.


Subject(s)
Antigens, Neoplasm/administration & dosage , Dendritic Cells/transplantation , Immune Checkpoint Inhibitors/administration & dosage , Immunotherapy/methods , Neoplasms/therapy , Oligodeoxyribonucleotides/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , Cancer Vaccines/administration & dosage , Combined Modality Therapy , Dose-Response Relationship, Drug , Female , Immunotherapy, Adoptive/methods , Liposomes , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Tumor Cells, Cultured
16.
Cancer Sci ; 112(9): 3469-3483, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34157192

ABSTRACT

Renal carcinoma shows a high risk of invasion and metastasis without effective treatment. Herein, we developed a chitosan (CS) nanoparticle-mediated DNA vaccine containing an activated factor L-Myc and a tumor-specific antigen CAIX for renal carcinoma treatment. The subcutaneous tumor models were intramuscularly immunized with CS-pL-Myc/pCAIX or control vaccine, respectively. Compared with single immunization group, the tumor growth was significantly suppressed in CS-pL-Myc/pCAIX co-immunization group. The increased proportion and mature of CD11c+ DCs, CD8+ CD11c+ DCs and CD103+ CD11c+ DCs were observed in the splenocytes from CS-pL-Myc/pCAIX co-immunized mice. Furthermore, the enhanced antigen-specific CD8+ T lymphocyte proliferation, cytotoxic T lymphocyte (CTL) responses, and multi-functional CD8+ T cell induction were detected in CS-pL-Myc/pCAIX co-immunization group compared with CS-pCAIX immunization group. Of note, the depletion of CD8 T cells resulted in the reduction of CD8+ T cells or CD8+ CD11c+ DCs and the loss of anti-tumor efficacy induced by CS-pL-Myc/pCAIX vaccine, suggesting the therapeutic efficacy of the vaccine was required for CD8+ DCs and CD103+ DCs mediated CD8+ T cells responses. Likewise, CS-pL-Myc/pCAIX co-immunization also significantly inhibited the lung metastasis of renal carcinoma models accompanied with the increased induction of multi-functional CD8+ T cell responses. Therefore, these results indicated that CS-pL-Myc/pCAIX vaccine could effectively induce CD8+ DCs and CD103+ DCs mediated tumor-specific multi-functional CD8+ T cell responses and exert the anti-tumor efficacy. This vaccine strategy offers a potential and promising approach for solid or metastatic tumor treatment.


Subject(s)
Antigens, CD/metabolism , Antigens, Neoplasm/administration & dosage , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Carbonic Anhydrase IX/administration & dosage , Carcinoma, Renal Cell/therapy , Chitosan/chemistry , Dendritic Cells/immunology , Drug Delivery Systems/methods , Immunity , Immunization/methods , Integrin alpha Chains/metabolism , Kidney Neoplasms/therapy , Nanoparticles/chemistry , Proto-Oncogene Proteins c-myc/administration & dosage , Vaccines, DNA/administration & dosage , Animals , Antigens, Neoplasm/genetics , Carbonic Anhydrase IX/genetics , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Disease Models, Animal , Female , HEK293 Cells , Humans , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-myc/genetics , Treatment Outcome , Vaccines, DNA/immunology
17.
Cancer Sci ; 112(9): 3484-3490, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34187084

ABSTRACT

For successful immunotherapy for cancer, it is important to understand the immunological status of tumor antigen-specific CD8+ T cells in the tumor microenvironment during tumor progression. In this study, we monitored the behavior of B16OVA-Luc cells in mice immunized with a model tumor antigen ovalbumin (OVA). Using bioluminescence imaging, we identified the time series of OVA-specific CD8+ T-cell responses during tumor progression: initial progression, immune control, and the escape phase. As a result of analyzing the status of tumor antigen-specific CD8+ cells in those 3 different phases, we found that the expression of NKG2D defines tumor-reacting effector CD8+ T cells. NKG2D may control the fate and TOX expression of tumor-reacting CD8+ T cells, considering that NKG2D blockade in OVA-vaccinated mice delayed the growth of the B16OVA-Luc2 tumor and increased the presence of tumor-infiltrating OVA-specific CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Tumor Microenvironment/immunology , Animals , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/metabolism , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Bromodeoxyuridine/administration & dosage , Bromodeoxyuridine/pharmacokinetics , Interferon-gamma/deficiency , Interferon-gamma/genetics , Luciferases/metabolism , Luminescent Measurements/methods , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage , Ovalbumin/metabolism , Skin Neoplasms/pathology , Vaccination/methods
18.
Nat Commun ; 12(1): 2626, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976179

ABSTRACT

By conferring systemic protection and durable benefits, cancer immunotherapies are emerging as long-term solutions for cancer treatment. One such approach that is currently undergoing clinical testing is a therapeutic anti-cancer vaccine that uses two different viruses expressing the same tumor antigen to prime and boost anti-tumor immunity. By providing the additional advantage of directly killing cancer cells, oncolytic viruses (OVs) constitute ideal platforms for such treatment strategy. However, given that the targeted tumor antigen is encoded into the viral genomes, its production requires robust infection and therefore, the vaccination efficiency partially depends on the unpredictable and highly variable intrinsic sensitivity of each tumor to OV infection. In this study, we demonstrate that anti-cancer vaccination using OVs (Adenovirus (Ad), Maraba virus (MRB), Vesicular stomatitis virus (VSV) and Vaccinia virus (VV)) co-administered with antigenic peptides is as efficient as antigen-engineered OVs and does not depend on viral replication. Our strategy is particularly attractive for personalized anti-cancer vaccines targeting patient-specific mutations. We suggest that the use of OVs as adjuvant platforms for therapeutic anti-cancer vaccination warrants testing for cancer treatment.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Disease Models, Animal , Female , Humans , Mice , Neoplasms/immunology , Oncolytic Viruses/genetics , Poly I-C/administration & dosage , Poly I-C/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccinia virus , Vesicular stomatitis Indiana virus , Xenograft Model Antitumor Assays
19.
Front Immunol ; 12: 627932, 2021.
Article in English | MEDLINE | ID: mdl-33859638

ABSTRACT

Vaccination was first pioneered in the 18th century by Edward Jenner and eventually led to the development of the smallpox vaccine and subsequently the eradication of smallpox. The impact of vaccination to prevent infectious diseases has been outstanding with many infections being prevented and a significant decrease in mortality worldwide. Cancer vaccines aim to clear active disease instead of aiming to prevent disease, the only exception being the recently approved vaccine that prevents cancers caused by the Human Papillomavirus. The development of therapeutic cancer vaccines has been disappointing with many early cancer vaccines that showed promise in preclinical models often failing to translate into efficacy in the clinic. In this review we provide an overview of the current vaccine platforms, adjuvants and delivery systems that are currently being investigated or have been approved. With the advent of immune checkpoint inhibitors, we also review the potential of these to be used with cancer vaccines to improve efficacy and help to overcome the immune suppressive tumor microenvironment.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Drug Delivery Systems , Gene Transfer Techniques , Neoplasms/therapy , Adjuvants, Immunologic/chemistry , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/chemistry , Cancer Vaccines/genetics , Drug Carriers , Drug Compounding , Humans , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Tumor Escape , Tumor Microenvironment , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , mRNA Vaccines
20.
Proc Natl Acad Sci U S A ; 117(50): 31780-31788, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33239441

ABSTRACT

Engineered cytokines are gaining importance in cancer therapy, but these products are often limited by toxicity, especially at early time points after intravenous administration. 4-1BB is a member of the tumor necrosis factor receptor superfamily, which has been considered as a target for therapeutic strategies with agonistic antibodies or using its cognate cytokine ligand, 4-1BBL. Here we describe the engineering of an antibody fusion protein, termed F8-4-1BBL, that does not exhibit cytokine activity in solution but regains biological activity on antigen binding. F8-4-1BBL bound specifically to its cognate antigen, the alternatively spliced EDA domain of fibronectin, and selectively localized to tumors in vivo, as evidenced by quantitative biodistribution experiments. The product promoted a potent antitumor activity in various mouse models of cancer without apparent toxicity at the doses used. F8-4-1BBL represents a prototype for antibody-cytokine fusion proteins, which conditionally display "activity on demand" properties at the site of disease on antigen binding and reduce toxicity to normal tissues.


Subject(s)
4-1BB Ligand/administration & dosage , Antigens, Neoplasm/administration & dosage , Neoplasms/drug therapy , Recombinant Fusion Proteins/administration & dosage , 4-1BB Ligand/genetics , 4-1BB Ligand/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Fibronectins/genetics , Fibronectins/immunology , Humans , Mice , Neoplasms/immunology , Protein Domains/genetics , Protein Domains/immunology , Protein Engineering , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL