Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
1.
J Med Chem ; 67(12): 9976-9990, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38886162

ABSTRACT

This study describes the design and synthesis of five TF-based cancer vaccine candidates using a lipid A mimetic as the carrier and a built-in adjuvant. All synthesized conjugates elicited robust and consistent TF-specific immune responses in mice without external adjuvants. Immunological studies subsequently conducted in wild-type and TLR4 knockout C57BL/6 mice demonstrated that the activation of TLR4 was the main reason that the synthesized lipid A mimetics increased the TF-specific immune responses. All antisera induced by these conjugates can specifically recognize, bind to, and induce the lysis of TF-positive cancer cells. Moreover, representative conjugates 2 and 3 could effectively reduce the growth of tumors and prolong the survival time of mice in vivo, and the efficacies were better than glycoprotein TF-CRM197 with alum adjuvant. Lipid A mimetics could therefore be a promising platform for the development of new carbohydrate-based vaccine carriers with self-adjuvanting properties for the treatment of cancer.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines , Drug Design , Lipid A , Mice, Inbred C57BL , Animals , Lipid A/analogs & derivatives , Lipid A/chemistry , Lipid A/pharmacology , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Cancer Vaccines/chemical synthesis , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/chemistry , Mice , Mice, Knockout , Humans , Female , Toll-Like Receptor 4/metabolism , Cell Line, Tumor
2.
ACS Appl Mater Interfaces ; 13(42): 49737-49753, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34648269

ABSTRACT

Peptide vaccines exhibit great potential in cancer therapy via eliciting antigen-specific host immune response and long-term immune memory to defend cancer cells. However, the low induced immune response of many developing vaccines implies the imperatives for understanding the favorable structural features of efficient cancer vaccines. Herein, we report on the two groups of self-adjuvanting peptide vaccines with distinct morphology and investigate the relationship between the morphology of peptide vaccines and the induced immune response. Two nanofibril peptide vaccines were created via co-assembly of a pentapeptide with a central 4-aminoproline residue, with its derivative functionalized with antigen epitopes derived from human papillomavirus E7 proteins, whereas utilization of a pentapeptide with a natural proline residue led to the formation of two nanoparticle peptide vaccines. The immunological results of dendritic cell (DCs) maturation and antigen presentation induced by the peptide assemblies implied the self-adjuvanting property of the resulting peptide vaccines. In particular, cellular uptake studies revealed the enhanced internalization and elongated retention of the nanofibril peptide vaccines in DCs, leading to their advanced performance in DC maturation, accumulation at lymph nodes, infiltration of cytotoxic T lymphocytes into tumor tissues, and eventually lysis of in vivo tumor cells, compared to the nanoparticle counterparts. The antitumor immune response caused by the nanofibril peptide vaccines was further augmented when simultaneously administrated with anti-PD-1 checkpoint blockades, suggesting the opportunity of the combinatorial immunotherapy by utilizing the nanofibril peptide vaccines. Our findings strongly demonstrate a robust relationship between the immune response of peptide vaccines and their morphology, thereby elucidating the critical role of morphological control in the design of efficient peptide vaccines and providing the guidance for the design of efficient peptide vaccines in the future.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cancer Vaccines/pharmacology , Oropharyngeal Neoplasms/therapy , Papillomaviridae/drug effects , Papillomavirus Infections/therapy , Vaccines, Subunit/pharmacology , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/chemistry , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Cancer Vaccines/chemical synthesis , Cancer Vaccines/chemistry , Cell Line , Humans , Immunotherapy , Materials Testing , Mice , Molecular Structure , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Oropharyngeal Neoplasms/immunology , Papillomaviridae/immunology , Papillomavirus Infections/immunology , Vaccines, Subunit/chemical synthesis , Vaccines, Subunit/chemistry
3.
J Med Chem ; 64(4): 1951-1965, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33539088

ABSTRACT

GM3, a typical tumor-associated carbohydrate antigen, is considered as an important target for cancer vaccine development, but its low immunogenicity limits its application. αGalCer, an iNKT cell agonist, has been employed as an adjuvant via a unique immune mode. Herein, we prepared and investigated two types of antitumor vaccine candidates: (a) self-adjuvanting vaccine GM3-αGalCer by conjugating GM3 with αGalCer and (b) noncovalent vaccine GM3-lipid/αGalCer, in which GM3 is linked with lipid anchor and coassembled with αGalCer. This demonstrated that ßGalCer is an exceptionally optimized lipid anchor, which enables the noncovalent vaccine candidate GM3-ßGalCer/αGalCer to evoke a comparable antibody level to GM3-αGalCer. However, the antibodies induced by GM3-αGalCer are better at recognition B16F10 cancer cells and more effectively activate the complement system. Our study highlights the importance of vaccine constructs utilizing covalent or noncovalent assembly between αGalCer with carbohydrate antigens and choosing an appropriate lipid anchor for use in noncovalent vaccine formulation.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cancer Vaccines/pharmacology , G(M3) Ganglioside/pharmacology , Galactosylceramides/pharmacology , Adjuvants, Immunologic/chemical synthesis , Animals , Cancer Vaccines/chemical synthesis , Cancer Vaccines/immunology , Carbohydrate Sequence , Female , G(M3) Ganglioside/analogs & derivatives , G(M3) Ganglioside/immunology , Galactosylceramides/chemical synthesis , Galactosylceramides/immunology , Humans , Immunity, Humoral/drug effects , Immunoglobulin G/immunology , Liposomes/chemistry , Mice, Inbred BALB C , Natural Killer T-Cells/immunology , THP-1 Cells
4.
J Med Chem ; 63(20): 11691-11706, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32960056

ABSTRACT

Self-adjuvanting vaccines, wherein an antigenic peptide is covalently bound to an immunostimulating agent, have been shown to be promising tools for immunotherapy. Synthetic Toll-like receptor (TLR) ligands are ideal adjuvants for covalent linking to peptides or proteins. We here introduce a conjugation-ready TLR4 ligand, CRX-527, a potent powerful lipid A analogue, in the generation of novel conjugate-vaccine modalities. Effective chemistry has been developed for the synthesis of the conjugation-ready ligand as well as the connection of it to the peptide antigen. Different linker systems and connection modes to a model peptide were explored, and in vitro evaluation of the conjugates showed them to be powerful immune-activating agents, significantly more effective than the separate components. Mounting the CRX-527 ligand at the N-terminus of the model peptide antigen delivered a vaccine modality that proved to be potent in activation of dendritic cells, in facilitating antigen presentation, and in initiating specific CD8+ T-cell-mediated killing of antigen-loaded target cells in vivo. Synthetic TLR4 ligands thus show great promise in potentiating the conjugate vaccine platform for application in cancer vaccination.


Subject(s)
Cancer Vaccines/chemical synthesis , Glucosamine/analogs & derivatives , Lipid A/analogs & derivatives , Organophosphorus Compounds/chemistry , Ovalbumin/chemistry , Toll-Like Receptor 4/immunology , Adjuvants, Immunologic , Animals , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cytokines/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Glucosamine/chemistry , Glucosamine/immunology , Immunoglobulin G/blood , Ligands , Lymphocyte Activation/drug effects , Mice , Organophosphorus Compounds/immunology , Ovalbumin/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Vaccines, Conjugate
5.
Cancer Biol Med ; 17(1): 132-141, 2020 02 15.
Article in English | MEDLINE | ID: mdl-32296581

ABSTRACT

Objective: Cancer stem cell is one of the important causes of tumorigenesis as well as a drug target in the treatment of malignant tumor. However, at present, there is no immune vaccine targeting these cells. Octamer-binding transcription factor 4 (OCT4), a marker of embryonic stem cells and germ cells, often highly expresses in the early stages of tumorigenesis and is therefore a good candidate for cancer vaccine development. Methods: To identify the optimal carrier and adjuvant combination, we chemically synthesized and linked three different OCT4 epitope antigens to a carrier protein, keyhole limpet hemocyanin (KLH), combined with Toll-like receptor 9 agonist (TLR9). Results: Immunization with OCT4-3 + TLR9 produced the strongest immune response in mice. In prevention assays, significant tumor growth inhibition was achieved in BABL/c mice treated with OCT4-3 + TLR9 (P < 0.01). Importantly, the results showed that cytotoxic T lymphocyte activity and the inhibition of tumor growth were enhanced in mice immunized with OCT4-3 combined with TLR9. Meanwhile, multiple cytokines [such as interferon (IFN)-γ (P < 0.05), interleukin (IL)-12 (P < 0.05), IL-2 (P < 0.01), and IL-6 (P < 0.05)] promoting cellular immune responses were shown to be greatly enhanced in mice immunized with OCT4-3 + TLR9. Moreover, we considered safety considerations in terms of the composition of the vaccines to help facilitate the development of effective next-generation vaccines. Conclusions: Collectively, these experiments demonstrated that combination therapy with TLR9 agonist induced a tumor-specific adaptive immune response, leading to the suppression of primary tumor growth in testis embryonic carcinoma.


Subject(s)
Cancer Vaccines/administration & dosage , Neoplasms/therapy , Neoplastic Stem Cells/immunology , Octamer Transcription Factor-3/immunology , Toll-Like Receptor 9/agonists , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/genetics , Animals , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/chemical synthesis , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Epitopes/administration & dosage , Epitopes/chemistry , Epitopes/immunology , Hemocyanins/administration & dosage , Hemocyanins/genetics , Hemocyanins/immunology , Humans , Immunogenicity, Vaccine , Male , Mice , Neoplasms/immunology , Neoplasms/pathology , Octamer Transcription Factor-3/genetics , Peptides/chemical synthesis , Peptides/genetics , Peptides/immunology , Toll-Like Receptor 9/metabolism , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
6.
ACS Chem Biol ; 15(3): 728-739, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32045202

ABSTRACT

Dendritic cells (DCs) are armed with a multitude of Pattern Recognition Receptors (PRRs) to recognize pathogens and initiate pathogen-tailored T cell responses. In these responses, the maturation of DCs is key, as well as the production of cytokines that help to accomplish T cell responses. DC-SIGN is a frequently exploited PRR that can effectively be targeted with mannosylated antigens to enhance the induction of antigen-specific T cells. The natural O-mannosidic linkage is susceptible to enzymatic degradation, and its chemical sensitivity complicates the synthesis of mannosylated antigens. For this reason, (oligo)mannosides are generally introduced in a late stage of the antigen synthesis, requiring orthogonal conjugation handles for their attachment. To increase the stability of the mannosides and streamline the synthesis of mannosylated peptide antigens, we here describe the development of an acid-stable C-mannosyl lysine, which allows for the inline introduction of mannosides during solid-phase peptide synthesis (SPPS). The developed amino acid has been successfully used for the assembly of both small ligands and peptide antigen conjugates comprising an epitope of the gp100 melanoma-associated antigen and a TLR7 agonist for DC activation. The ligands showed similar internalization capacities and binding affinities as the O-mannosyl analogs. Moreover, the antigen conjugates were capable of inducing maturation, stimulating the secretion of pro-inflammatory cytokines, and providing enhanced gp100 presentation to CD8+ and CD4+ T cells, similar to their O-mannosyl counterparts. Our results demonstrate that the C-mannose lysine is a valuable building block for the generation of anticancer peptide-conjugate vaccine modalities.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cancer Vaccines/chemical synthesis , Glycopeptides/chemistry , Lysine/chemistry , Mannose/chemistry , Vaccines, Conjugate/chemistry , Antigens, Neoplasm/metabolism , Antineoplastic Agents/metabolism , Cancer Vaccines/metabolism , Cell Culture Techniques , Cytokines/metabolism , Dendritic Cells , Epitopes/chemistry , Epitopes/metabolism , Fluorescent Dyes/chemistry , Humans , Optical Imaging , T-Lymphocytes , Toll-Like Receptor 7/metabolism , Vaccines, Conjugate/metabolism , Vaccines, Synthetic/chemistry , gp100 Melanoma Antigen/metabolism
7.
Chem Commun (Camb) ; 55(91): 13741-13744, 2019 Nov 12.
Article in English | MEDLINE | ID: mdl-31661093

ABSTRACT

Natural glycopeptides have been shown to possess interesting biological activities. In this work, we have developed a general solid-phase approach to C-terminal glycopeptides. Taking advantage of oxime resin ester bond nucleophile susceptibility, we optimised the nucleophilic cleavage step with glycosylamines and demonstrated the generality and scope of this method. In addition, this reaction has high functional group tolerance and can be used for the preparation of longer C-terminal glycopeptides, demonstrated with the synthesis of a glycododecapeptide in one single step. The results pave the way to access efficiently novel medically relevant compounds.


Subject(s)
Glycopeptides/chemistry , Oximes/chemistry , Antigens, Tumor-Associated, Carbohydrate/chemistry , Biomarkers, Tumor/chemical synthesis , Biomarkers, Tumor/chemistry , Cancer Vaccines/chemical synthesis , Cancer Vaccines/chemistry , Glycopeptides/chemical synthesis , Glycosylation , Humans
8.
ACS Chem Biol ; 14(10): 2176-2184, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31498587

ABSTRACT

Mucin-1 (MUC1) is a highly attractive antigenic target for anticancer vaccines. Naturally existing MUC1 can contain multiple types of O-linked glycans, including the Thomsen-Friedenreich (Tf) antigen and the Sialyl Thomsen-nouveau (STn) antigen. In order to target these antigens as potential anticancer vaccines, MUC1 glycopeptides SAPDT*RPAP (T* is the glycosylation site) bearing the Tf and the STn antigen, respectively, have been synthesized. The bacteriophage Qß carrier is a powerful carrier for antigen delivery. The conjugates of MUC1-Tf and -STn glycopeptides with Qß were utilized to immunize immune-tolerant human MUC1 transgenic (MUC1.Tg) mice, which elicited superior levels of anti-MUC1 IgG antibodies with titers reaching over 2 million units. The IgG antibodies recognized a wide range of MUC1 glycopeptides bearing diverse glycans. Antibodies induced by Qß-MUC1-Tf showed strongest binding, with MUC1-expressing melanoma B16-MUC1 cells, and effectively killed these cells in vitro. Vaccination with Qß-MUC1-Tf first followed by tumor challenge in a lung metastasis model showed significant reductions of the number of tumor foci in the lungs of immunized mice as compared to those in control mice. This was the first time that a MUC1-Tf-based vaccine has shown in vivo efficacy in a tumor model. As such, Qß-MUC1 glycopeptide conjugates have great potential as anticancer vaccines.


Subject(s)
Cancer Vaccines/therapeutic use , Glycopeptides/therapeutic use , Immunoconjugates/therapeutic use , Mucin-1/immunology , Peptide Fragments/therapeutic use , Viral Proteins/therapeutic use , Allolevivirus/chemistry , Amino Acid Sequence , Animals , Antigens, Tumor-Associated, Carbohydrate/immunology , Cancer Vaccines/chemical synthesis , Cancer Vaccines/immunology , Cell Line, Tumor , Female , Glycopeptides/chemical synthesis , Glycopeptides/immunology , Humans , Immunoconjugates/immunology , Immunoglobulin G/immunology , Lung Neoplasms/therapy , Male , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/chemical synthesis , Peptide Fragments/immunology , Viral Proteins/chemical synthesis , Viral Proteins/immunology
9.
Glycoconj J ; 36(5): 399-408, 2019 10.
Article in English | MEDLINE | ID: mdl-31267246

ABSTRACT

Even though a vaccine that targets tumor-associated carbohydrate antigens on epithelial carcinoma cells presents an attractive therapeutic approach, relatively poor immunogenicity limits its development. In this study, we investigated the immunological activity of a fluoro-substituted Sialyl-Tn (F-STn) analogue coupled to the non-toxic cross-reactive material of diphtheria toxin197 (CRM197). Our results indicate that F-STn-CRM197 promotes a greater immunogenicity than non-fluorinated STn-CRM197. In the presence or absence of adjuvant, F-STn-CRM197 remarkably enhances both cellular and humoral immunity against STn by increasing antigen-specific lymphocyte proliferation and inducing a mixed Th1/Th2 response leading to production of IFN-γ and IL-4 cytokines, as well as STn-specific antibodies. Furthermore, antisera produced from F-STn-CRM197 immunization significantly recognizes STn-positive tumor cells and increases cancer cell lysis induced by antibody-dependent cell-mediated cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) pathways. Our data suggest that this F-STn vaccine may be useful for cancer immunotherapy and possibly for prophylactic prevention of cancer.


Subject(s)
Antibodies, Neoplasm/pharmacology , Antigens, Tumor-Associated, Carbohydrate/chemistry , Bacterial Proteins/pharmacology , Cancer Vaccines/pharmacology , Colonic Neoplasms/therapy , Glycoconjugates/pharmacology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Neoplasm/isolation & purification , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, Tumor-Associated, Carbohydrate/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Cancer Vaccines/chemical synthesis , Cancer Vaccines/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Female , Gene Expression , Glycoconjugates/chemical synthesis , Glycoconjugates/immunology , Halogenation , Humans , Immune Sera/chemistry , Immune Sera/pharmacology , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Immunization , Immunogenicity, Vaccine , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Lymphocytes/drug effects , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Spleen/drug effects , Spleen/immunology , Th1-Th2 Balance
10.
Nat Commun ; 10(1): 1899, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015397

ABSTRACT

Nanoparticles can potentially stimulate tumour microenvironments to elicit antitumour immunity. Herein, we demonstrate effective immunotherapy of colorectal cancer via systemic delivery of an immunostimulatory chemotherapeutic combination in nanoscale coordination polymer (NCP) core-shell particles. Oxaliplatin and dihydroartemesinin have contrasting physicochemical properties but strong synergy in reactive oxygen species (ROS) generation and anticancer activity. The combined ROS generation is harnessed for immune activation to synergize with an anti-PD-L1 antibody for the treatment of murine colorectal cancer tumours. The favourable biodistribution and tumour uptake of NCPs and the absence of peripheral neuropathy allow for repeated dosing to afford 100% tumour eradication. The involvement of innate and adaptive immune systems elicit strong and long lasting antitumour immunity which prevents tumour formation when cured mice are challenged with cancer cells. The intrinsically biodegradable, well tolerated, and systemically available immunostimulatory NCP promises to enter clinical testing as an immunotherapy against colorectal cancer.


Subject(s)
Adenocarcinoma/therapy , Cancer Vaccines/pharmacology , Colorectal Neoplasms/therapy , Immunologic Factors/pharmacology , Nanoparticles/administration & dosage , Polymers/administration & dosage , Adaptive Immunity/drug effects , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/mortality , Animals , Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Artemisinins/pharmacokinetics , Artemisinins/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cancer Vaccines/chemical synthesis , Cancer Vaccines/pharmacokinetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Drug Compounding/methods , Humans , Immunity, Innate/drug effects , Immunologic Factors/chemical synthesis , Immunologic Factors/pharmacokinetics , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Neoplasm Transplantation , Oxaliplatin/pharmacokinetics , Oxaliplatin/pharmacology , Polymers/chemical synthesis , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/agonists , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Survival Analysis , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
11.
Cancer Immunol Res ; 7(2): 174-182, 2019 02.
Article in English | MEDLINE | ID: mdl-30679156

ABSTRACT

T-cell recognition of cancer neoantigens is important for effective immune-checkpoint blockade therapy, and an increasing interest exists in developing personalized tumor neoantigen vaccines. Previous studies utilizing RNA and long-peptide neoantigen vaccines in preclinical and early-phase clinical studies have shown immune responses predominantly driven by MHC class II CD4+ T cells. Here, we report on a preclinical study utilizing a DNA vaccine platform to target tumor neoantigens. We showed that optimized strings of tumor neoantigens, when delivered by potent electroporation-mediated DNA delivery, were immunogenic and generated predominantly MHC class I-restricted, CD8+ T-cell responses. High MHC class I affinity was associated specifically with immunogenic CD8+ T-cell epitopes. These DNA neoantigen vaccines induced a therapeutic antitumor response in vivo, and neoantigen-specific T cells expanded from immunized mice directly killed tumor cells ex vivo These data illustrate a unique advantage of this DNA platform to drive CD8+ T-cell immunity for neoantigen immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Vaccines, DNA/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/chemical synthesis , Cytotoxicity, Immunologic , Melanoma, Experimental , Mice , Neoplasms/immunology , Neoplasms/therapy , Vaccines, DNA/chemical synthesis , Vaccinology/methods
12.
Curr Med Chem ; 26(3): 427-445, 2019.
Article in English | MEDLINE | ID: mdl-29376489

ABSTRACT

Current advances in cancer treatment are based on the recent discoveries of molecular mechanisms of tumour maintenance. It was shown that heat shock proteins (HSPs) play a crucial role in the development of immune response against tumours. Thus, HSPs represent multifunctional agents not only with chaperone functions, but also possessing immunomodulatory properties. These properties are exploited for the development of HSP-based anticancer vaccines aimed to induce cytotoxic responses against tumours. To date, a number of strategies have been suggested to facilitate HSP-based vaccine production and to increase its effectiveness. The present review focuses on the current trend for the development of HSPbased vaccines aimed at inducing strong immunological tumour-specific responses against cancer cells of distinct etiology and localization.


Subject(s)
Cancer Vaccines/chemical synthesis , Heat-Shock Proteins/immunology , Animals , Antigens, Neoplasm/immunology , Heat-Shock Proteins/chemical synthesis , Humans
13.
Chem Commun (Camb) ; 55(2): 253-256, 2018 Dec 20.
Article in English | MEDLINE | ID: mdl-30534737

ABSTRACT

We describe the preparation of a cancer vaccine candidate by conjugating a MUC1 peptide antigen to the ß-glucan polysaccharide, which serves both as a carrier and an immune activator. In contrast to amorphous polysaccharides, peptide-ß-glucan conjugates form uniform nanoparticles that facilitate the delivery of antigens and binding to myeloid cells, thus leading to the activation of both innate and adaptive immunity.


Subject(s)
Adenocarcinoma/immunology , Cancer Vaccines/immunology , Drug Carriers/chemistry , Mucin-1/immunology , Peptide Fragments/immunology , beta-Glucans/chemistry , Adaptive Immunity/immunology , Amino Acid Sequence , Animals , Cancer Vaccines/chemical synthesis , Cancer Vaccines/chemistry , Humans , Immunity, Active/immunology , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Interferon-gamma/metabolism , Interleukin-6/metabolism , MCF-7 Cells , Mice, Inbred C57BL , Mucin-1/chemistry , Peptide Fragments/chemistry , Vaccines, Subunit/chemical synthesis , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology
14.
ACS Chem Biol ; 13(12): 3279-3285, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30359529

ABSTRACT

Access to lipopeptide-based vaccines for immunological studies remains a significant challenge owing to the amphipathic nature of the molecules, which makes them difficult to synthesize and purify to homogeneity. Here, we describe the application of a new peptide ligation technology, the diselenide-selenoester ligation (DSL), to access self-adjuvanting glycolipopeptide vaccines. We show that rapid ligation of glyco- and lipopeptides is possible via DSL in mixed organic solvent-aqueous buffer and, when coupled with deselenization chemistry, affords rapid and efficient access to a vaccine candidate possessing a MUC1 glycopeptide epitope and the lipopeptide adjuvant Pam2Cys. This construct was shown to elicit MUC1-specific antibody and cytotoxic T lymphocyte responses in the absence of any other injected lipids or adjuvants. The inclusion of the helper T cell epitope PADRE both boosted the antibody response and resulted in elevated cytokine production.


Subject(s)
Adjuvants, Immunologic/chemical synthesis , Cancer Vaccines/immunology , Glycopeptides/immunology , Lipopeptides/immunology , Mucin-1/immunology , Organoselenium Compounds/chemistry , Amino Acid Sequence , Animals , Cancer Vaccines/chemical synthesis , Chemistry Techniques, Synthetic/methods , Female , Glycopeptides/chemical synthesis , Humans , Lipopeptides/chemical synthesis , MCF-7 Cells , Mice, Inbred C57BL , Minisatellite Repeats , Mucin-1/genetics , Organoselenium Compounds/chemical synthesis
15.
PLoS One ; 13(7): e0199249, 2018.
Article in English | MEDLINE | ID: mdl-29969453

ABSTRACT

Recently, many autologous tumor antigens have been examined for their potential use in cancer immunotherapy. However, the success of cancer vaccines in clinical trials has been limited, partly because of the limitations of using single, short peptides in most attempts. With this in mind, we aimed to develop multivalent synthetic long peptide (SLP) vaccines containing multiple cytotoxic T-lymphocyte (CTL) epitopes. However, to confirm whether a multivalent vaccine can induce an individual epitope-specific CTL, the only viable screening strategies currently available are interferon-gamma (IFN-γ enzyme-linked immunospot (ELISPOT) assays using human peripheral blood mononuclear cells, or expensive human leukocyte antigen (HLA)-expressing mice. In this report, we evaluated the use of our developed murine-20S immunoproteasome (i20S) digestion assay, and found that it could predict the results of IFN-γ ELISPOT assays. Importantly, the murine-i20S digestion assay not only predicted CTL induction, but also antitumor activity in an HLA-expressing mouse model. We conclude that the murine-i20S digestion assay is an extremely useful tool for the development of "all functional" multivalent SLP vaccines.


Subject(s)
Cancer Vaccines/pharmacology , HLA-A2 Antigen/genetics , Immunoassay , Immunotherapy, Active/methods , Melanoma, Experimental/prevention & control , Peptides/pharmacology , Amino Acid Sequence , Animals , Cancer Vaccines/chemical synthesis , Cancer Vaccines/immunology , Enzyme-Linked Immunospot Assay , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lymphocyte Activation/drug effects , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Transgenic , Peptides/chemical synthesis , Peptides/immunology , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Transgenes , Tumor Burden/drug effects , Vaccines, Subunit
16.
Angew Chem Int Ed Engl ; 57(27): 8219-8224, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29768704

ABSTRACT

Sialyl-Tn (STn) is a tumor-associated carbohydrate antigen (TACA) rarely observed on healthy tissues. We synthesized two fully synthetic N-acetyl and N-propionyl STn trimer (triSTn) vaccines possessing a T-helper epitope and a TLR2 agonist, since the clustered STn antigens are highly expressed on many cancer cells. Immunization of both vaccines in mice induced the anti-triSTn IgG antibodies, which recognized triSTn-expressing cell lines PANC-1 and HepG2. The N-propionyl triSTn vaccine induced the triSTn-specific IgGs, while IgGs induced by the N-acetyl triSTn vaccine were less specific. These results illustrated that N-propionyl triSTn is a valuable unnatural TACA for anticancer vaccines.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/immunology , Cancer Vaccines/immunology , Epitopes/immunology , Animals , Antibodies, Neoplasm/immunology , Antigens, Tumor-Associated, Carbohydrate/chemistry , Cancer Vaccines/chemical synthesis , Cattle , Cell Line, Tumor , Epitopes/chemistry , Hep G2 Cells , Humans , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Serum Albumin, Bovine/chemistry , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism
17.
ACS Chem Biol ; 13(6): 1668-1676, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29782143

ABSTRACT

Mucin-1 (MUC1) is one of the top ranked tumor associated antigens. In order to generate effective anti-MUC1 immune responses as potential anticancer vaccines, MUC1 peptides and glycopeptides have been covalently conjugated to bacteriophage Qß. Immunization of mice with these constructs led to highly potent antibody responses with IgG titers over one million, which are among the highest anti-MUC1 IgG titers reported to date. Furthermore, the high IgG antibody levels persisted for more than six months. The constructs also elicited MUC1 specific cytotoxic T cells, which can selectively kill MUC1 positive tumor cells. The unique abilities of Qß-MUC1 conjugates to powerfully induce both antibody and cytotoxic T cell immunity targeting tumor cells bode well for future translation of the constructs as anticancer vaccines.


Subject(s)
Bacteriophages/immunology , Cancer Vaccines/immunology , Immunity, Humoral/immunology , Immunoglobulin G/immunology , Lymphocyte Activation/immunology , Mucin-1/immunology , Amino Acid Sequence , Animals , Antibody Formation/immunology , Bacteriophages/chemistry , Cancer Vaccines/chemical synthesis , Cell Line, Tumor , Humans , Immunization , Lymphoma/immunology , Mice, Inbred C57BL , Microarray Analysis , Mucin-1/chemistry , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology
18.
Nat Protoc ; 13(2): 335-357, 2018 02.
Article in English | MEDLINE | ID: mdl-29345636

ABSTRACT

This protocol describes how to induce large numbers of tumor-specific cytotoxic T cells (CTLs) in the spleens and lymph nodes of mice receiving dendritic cell (DC) vaccines and how to modulate tumor microenvironments (TMEs) to ensure effective homing of the vaccination-induced CTLs to tumor tissues. We also describe how to evaluate the numbers of tumor-specific CTLs within tumors. The protocol contains detailed information describing how to generate a specialized DC vaccine with augmented ability to induce tumor-specific CTLs. We also describe methods to modulate the production of chemokines in the TME and show how to quantify tumor-specific CTLs in the lymphoid organs and tumor tissues of mice receiving different treatments. The combined experimental procedure, including tumor implantation, DC vaccine generation, chemokine-modulating (CKM) approaches, and the analyses of tumor-specific systemic and intratumoral immunity is performed over 30-40 d. The presented ELISpot-based ex vivo CTL assay takes 6 h to set up and 5 h to develop. In contrast to other methods of evaluating tumor-specific immunity in tumor tissues, our approach allows detection of intratumoral T-cell responses to nonmanipulated weakly immunogenic cancers. This detection method can be performed using basic laboratory skills, and facilitates the development and preclinical evaluation of new immunotherapies.


Subject(s)
Dendritic Cells/physiology , Immunotherapy/methods , T-Lymphocytes, Cytotoxic/immunology , Animals , Cancer Vaccines/chemical synthesis , Cancer Vaccines/metabolism , Cell Line , Chemokines , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Neoplasms , Spleen/immunology , T-Lymphocytes, Cytotoxic/physiology , Tumor Microenvironment/physiology
19.
Org Biomol Chem ; 16(6): 993-999, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29345713

ABSTRACT

Therapeutic vaccines have been regarded as a very promising treatment modality against cancer. Tumor-associated MUC1 is a promising antigen for the design of antitumor vaccines. However, body's immune tolerance and low immunogenicity of MUC1 glycopeptides limited their use as effective antigen epitopes of therapeutic vaccines. To solve this problem, we chose the immune dominant region of MUC1 VNTRs. We designed and synthesized its linear trivalent glycopeptide fragments and coupled the fragments with BSA. Immunological evaluation indicated that the antibodies induced by glycosylated MUC1 based vaccine 11 had a stronger binding than non-glycosylated 10. The novel constructed antigen epitopes have the potential to overcome the weak immunogenicity of natural MUC1 glycopeptides and deserve further research.


Subject(s)
Cancer Vaccines/immunology , Glycopeptides/immunology , Mucin-1/immunology , Peptide Fragments/immunology , Serum Albumin, Bovine/immunology , Adenocarcinoma/immunology , Animals , Breast Neoplasms/immunology , Cancer Vaccines/chemical synthesis , Female , Glycopeptides/chemical synthesis , Humans , Immunodominant Epitopes , Immunogenicity, Vaccine/immunology , MCF-7 Cells , Mice, Inbred BALB C , Mucin-1/chemistry , Peptide Fragments/chemical synthesis , Serum Albumin, Bovine/chemical synthesis , Tandem Repeat Sequences , Vaccines, Subunit/chemical synthesis , Vaccines, Subunit/immunology
20.
Chembiochem ; 19(2): 121-125, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29120508

ABSTRACT

A fully synthetic MUC1-based cancer vaccine was designed and chemically synthesized containing an endogenous helper T-epitope (MHC class II epitope). The vaccine elicited robust IgG titers that could neutralize cancer cells by antibody-dependent cell-mediated cytotoxicity (ADCC). It also activated cytotoxic T-lymphocytes. Collectively, the immunological data demonstrate engagement of helper T-cells in immune activation. A synthetic methodology was developed for a penta-glycosylated MUC1 glycopeptide, and antisera of mice immunized by the new vaccine recognized such a structure. Previously reported fully synthetic MUC1-based cancer vaccines that elicited potent immune responses employed exogenous helper T-epitopes derived from microbes. It is the expectation that the use of the newly identified endogenous helper T-epitope will be more attractive, because it will activate cognate CD4+ T-cells that will provide critical tumor-specific help intratumorally during the effector stage of tumor rejection and will aid in the generation of sustained immunological memory.


Subject(s)
Cancer Vaccines/chemical synthesis , Cancer Vaccines/immunology , Glycopeptides/immunology , Mucin-1/immunology , Vaccines, Synthetic/immunology , Animals , Cancer Vaccines/chemistry , Glycopeptides/chemistry , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Conformation , Mucin-1/chemistry , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccines, Synthetic/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL