Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 503
Filtrar
1.
ACS Nano ; 18(19): 12386-12400, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38699808

RESUMO

Current cancer vaccines face challenges due to an immunosuppressive tumor microenvironment and their limited ability to produce an effective immune response. To address the above limitations, we develop a 3-(2-spiroadamantyl)-4-methoxy-4-(3-phosphoryloxy)-phenyl-1,2-dioxetane (alkaline phosphatase substrate) and XMD8-92 (extracellular signal-regulated kinase 5 inhibitor)-codelivered copper-tetrahydroxybenzoquinone (Cu-THBQ/AX) nanosized metal-organic framework to in situ-generate therapeutic vaccination. Once inside the early endosome, the alkaline phosphatase overexpressed in the tumor cells' membrane activates the in situ type I photodynamic effect of Cu-THBQ/AX for generating •O2-, and the Cu-THBQ/AX catalyzes O2 and H2O2 to •O2- and •OH via semiquinone radical catalysis and Fenton-like reactions. This surge of ROS in early endosomes triggers caspase-3-mediated proinflammatory pyroptosis via activating phospholipase C. Meanwhile, Cu-THBQ/AX can also induce the oligomerization of dihydrolipoamide S-acetyltransferase to trigger tumor cell cuproptosis. The production of •OH could also trigger the release of XMD8-92 for effectively inhibiting the efferocytosis of macrophages to convert immunosuppressive apoptosis of cancer cells into proinflammatory secondary necrosis. The simultaneous induction of pyroptosis, cuproptosis, and secondary necrosis effectively converts the tumor microenvironment from "cold" to "hot" conditions, making it an effective antigen pool. This transformation successfully activates the antitumor immune response, inhibiting tumor growth and metastasis.


Assuntos
Vacinas Anticâncer , Cobre , Macrófagos , Estruturas Metalorgânicas , Piroptose , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Animais , Camundongos , Piroptose/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Humanos , Cobre/química , Cobre/farmacologia , Vacinas Anticâncer/química , Microambiente Tumoral/efeitos dos fármacos , Nanopartículas/química , Fagocitose/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/química , Linhagem Celular Tumoral , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neoplasias/metabolismo , Camundongos Endogâmicos BALB C , Eferocitose , Nanovacinas
2.
J Med Chem ; 67(8): 6822-6838, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38588468

RESUMO

Weak antigens represented by MUC1 are poorly immunogenic, which greatly constrains the development of relevant vaccines. Herein, we developed a multifunctional lipidated protein as a carrier, in which the TLR1/2 agonist Pam3CSK4 was conjugated to the N-terminus of MUC1-loaded carrier protein BSA through pyridoxal 5'-phosphate-mediated transamination reaction. The resulting Pam3CSK4-BSA-MUC1 conjugate was subsequently incorporated into liposomes, which biomimics the membrane structure of tumor cells. The results indicated that this lipidated protein carrier significantly enhanced antigen uptake by APCs and obviously augmented the retention of the vaccine at the injection site. Compared with the BSA-MUC1 and BSA-MUC1 + Pam3CSK4 groups, Pam3CSK4-BSA-MUC1 evoked 22- and 11-fold increases in MUC1-specific IgG titers. Importantly, Pam3CSK4-BSA-MUC1 elicited robust cellular immunity and significantly inhibited tumor growth. This is the first time that lipidated protein was constructed to enhance antigen immunogenicity, and this universal carrier platform exhibits promise for utilization in various vaccines, holding the potential for further clinical application.


Assuntos
Lipossomos , Mucina-1 , Animais , Mucina-1/imunologia , Mucina-1/química , Camundongos , Humanos , Lipopeptídeos/química , Lipopeptídeos/imunologia , Lipopeptídeos/farmacologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/química , Soroalbumina Bovina/química , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/química , Feminino , Camundongos Endogâmicos BALB C , Antígenos/imunologia , Linhagem Celular Tumoral
3.
J Am Chem Soc ; 146(17): 11679-11693, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38482849

RESUMO

Lipid nanoparticles (LNPs)-based messenger RNA (mRNA) therapeutics have emerged with promising potentials in the fields of infectious diseases, cancer vaccines, and protein replacement therapies; however, their therapeutic efficacy and safety can still be promoted by the optimization of LNPs formulations. Unfortunately, current LNPs suffer from increased production of reactive oxygen species during translation, which leads to a decreased translation efficiency and the onset of inflammation and other side effects. Herein, we synthesize a lipid-modified poly(guanidine thioctic acid) polymer to fabricate novel LNPs for mRNA vaccines. The acquired G-LNPs significantly promote the translation efficiency of loaded mRNA and attenuate inflammation after vaccination through the elimination of reactive oxygen species that are responsible for translational inhibition and inflammatory responses. In vivo studies demonstrate the excellent antitumor efficacy of the G-LNPs@mRNA vaccine, and two-dose vaccination dramatically increases the population and infiltration of cytotoxic T cells due to the intense antitumor immune responses, thus generating superior antitumor outcomes compared with the mRNA vaccine prepared from traditional LNPs. By synergy with immune checkpoint blockade, the tumor inhibition of G-LNPs@mRNA is further boosted, indicating that G-LNPs-based mRNA vaccines will be powerful and versatile platforms to combat cancer.


Assuntos
Vacinas Anticâncer , Lipídeos , Lipossomos , Nanopartículas , RNA Mensageiro , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Nanopartículas/química , Animais , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Lipídeos/química , Humanos , Ácido Tióctico/química , Ácido Tióctico/farmacologia , Polímeros/química , Guanidinas/química , Guanidinas/farmacologia , Linhagem Celular Tumoral
4.
Adv Mater ; 36(19): e2309927, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38387609

RESUMO

Cytokines are powerful in cancer immunotherapy, however, their therapeutic potential is limited by the severe systemic toxicity. Here a potent strategy to reduce the toxicity of systemic cytokine therapy by delivering its denatured form using a finely designed nanochaperone, is described. It is demonstrated that even if the denatured protein cargos are occasionally released under normal physiological conditions they are still misfolded, while can effectively refold into native states and release to function in tumor microenvironment. Consequently, the systemic toxicity of cytokines is nearly completely overcome. Moreover, an immunogenic cell death (ICD)-inducing chemotherapeutic is further loaded and delivered to tumor using this nanochaperone to trigger the release of tumor-associated antigens (TAAs) that are subsequently captured in situ by nanochaperone and then reflows into lymph nodes (LNs) to promote antigen cross-presentation. This optimized personalized nanochaperone-vaccine demonstrates unprecedented suppressive effects against large, advanced tumors, and in combination with immune checkpoint blockade (ICB) therapy results in a significant abscopal effect and inhibition of postoperative tumor recurrence and metastasis. Hence, this approach provides a simple and universal delivery strategy to reduce the systemic toxicities of cytokines, as well as provides a robust personalized cancer vaccination platform, which may find wide applications in cancer immunotherapy.


Assuntos
Antígenos de Neoplasias , Imunoterapia , Interleucina-12 , Animais , Interleucina-12/metabolismo , Camundongos , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Linhagem Celular Tumoral , Humanos , Dobramento de Proteína , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Microambiente Tumoral/efeitos dos fármacos , Vacinas Anticâncer/química , Nanopartículas/química , Morte Celular Imunogênica/efeitos dos fármacos , Nanoestruturas/química
5.
Angew Chem Int Ed Engl ; 62(47): e202309744, 2023 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-37781858

RESUMO

Sialyl Lewisa (sLea ), also known as cancer antigen 19-9 (CA19-9), is a tumor-associated carbohydrate antigen. The overexpression of sLea on the surface of a variety of cancer cells makes it an attractive target for anticancer immunotherapy. However, sLea -based anticancer vaccines have been under-explored. To develop a new vaccine, efficient stereoselective synthesis of sLea with an amine-bearing linker was achieved, which was subsequently conjugated with a powerful carrier bacteriophage, Qß. Mouse immunization with the Qß-sLea conjugate generated strong and long-lasting anti-sLea IgG antibody responses, which were superior to those induced by the corresponding conjugate of sLea with the benchmark carrier keyhole limpet hemocyanin. Antibodies elicited by Qß-sLea were highly selective toward the sLea structure, could bind strongly with sLea -expressing cancer cells and human pancreatic cancer tissues, and kill tumor cells through complement-mediated cytotoxicity. Furthermore, vaccination with Qß-sLea significantly reduced tumor development in a metastatic cancer model in mice, demonstrating tumor protection for the first time by a sLea -based vaccine, thus highlighting the significant potential of sLea as a promising cancer antigen.


Assuntos
Bacteriófagos , Vacinas Anticâncer , Neoplasias , Camundongos , Humanos , Animais , Antígeno CA-19-9 , Vacinas Anticâncer/química , Imunoglobulina G/metabolismo
6.
Chem Soc Rev ; 52(10): 3353-3396, 2023 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-37070256

RESUMO

This review highlights the recent development in the use of carriers of increasing simplicities and versatile chemical ligation processes leading to synthetic vaccine candidates against tumor-associated carbohydrate antigens (TACAs). After briefly covering their structures, functions, occurrence, and biosynthesis, an overview of common conjugation chemistry is described with an emphasis on the versatile alkenyl glycosides as starting materials toward glycoconjugate syntheses. This is followed by a successive description of the numerous scaffolds and carriers used to progressively improve and simplify glycovaccine formulations. Throughout a systematic investigation of the various architectures involved, a critical description of the basic principles discovered en route to effective immune responses is disclosed wherein it is found that size, shape, densities, and carriers are all key factors involved towards successful vaccines.


Assuntos
Vacinas Anticâncer , Vacinas Anticâncer/química , Antígenos Glicosídicos Associados a Tumores/química , Vacinas Sintéticas/química , Glicoconjugados/química , Glicosídeos
7.
Small Methods ; 7(5): e2201409, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36802205

RESUMO

Adjuvants play an important role in enhancing vaccine-induced immune protection. Adequate cellular uptake, robust lysosomal escape, and subsequent antigen cross-presentation are critical steps for vaccine adjuvants to effectively elicit cellular immunity. Here, a fluorinated supramolecular strategy to generate a series of peptide adjuvants by using arginine (R) and fluorinated diphenylalanine peptide (DP) is adopted. It is found that the self-assembly ability and antigen-binding affinity of these adjuvants increase with the number of fluorine (F) and can be regulated by R. By comparison, 4RDP(F5) shows the strongest binding affinity with model antigen ovalbumin (OVA) and the best performance in dendritic cells maturation and antigen's lysosomal escape, which contributes to the subsequent antigen cross-presentation. As a consequence, 4RDP(F5)-OVA nanovaccine generates a strong cellular immunity in a prophylactic OVA-expressing EG7-OVA lymphoma model, leading to long-term immune memory for resisting tumor challenge. What's more, 4RDP(F5)-OVA nanovaccine in combination with anti-programmed cell death ligand-1 (anti-PD-L1) checkpoint blockade could effectively elicit anti-tumor immune responses and inhibit tumor growth in a therapeutic EG7-OVA lymphoma model. Overall, this study demonstrates the simplicity and effectiveness of fluorinated supramolecular strategies for constructing adjuvants and might provide an attractive vaccine adjuvant candidate for cancer immunotherapy.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , Vacinas Anticâncer/química , Vacinas Anticâncer/farmacologia , Apresentação de Antígeno , Adjuvantes Imunológicos , Antígenos , Neoplasias/terapia , Ovalbumina/química
8.
Acta Biomater ; 158: 535-546, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36632876

RESUMO

Vaccination shows great promise in cancer immunotherapy. However, the induction of robust and broad therapeutic CD8 T cell immunity against tumors is challenging due to the essential heterogenicity of tumor antigen expression. Recently, bioinspired materials have reshaped the field of cancer nanomedicine. Herein, a bioinspired nanofibrous trivalent peptide hydrogel vaccine was constructed using the spontaneous supramolecular co-assembly of three antigenic epitope-conjugated peptides, which could mimic the fibrillar structure and biological function of the extracellular matrix and naturally occurring protein assembly. The hydrogel vaccine could be accurately and flexibly adjusted to load each antigenic peptide at a defined ratio, which facilitated the antigen presentation of dendritic cells and significantly improved the initiation of CD8 T cell response and the secretion of interferon-γ (IFN-γ). C57BL/6 mice were immunized with the trivalent peptide hydrogel vaccine, where it elicited a high broad-spectrum antitumor CD8 T cell response that significantly inhibited the growth of B16 tumors in the absence of additional immunoadjuvants or delivery systems. In summary, the supramolecular assembly of triple antigenic epitope-conjugated peptides offers a simple, customizable, and versatile approach for the development of cancer vaccines with remarkable therapeutic efficacy, thereby providing a highly versatile platform for the application of personalized multivalent tumor vaccines. STATEMENT OF SIGNIFICANCE: (1) We report a feasible, versatile and bioinspired approach to manufacture a multivalent peptide-based hydrogel cancer vaccine in the absence of additional adjuvants, which closely mimics immune niches, co-delivers antigen epitopes, greatly promotes antigen presentation to DCs and their subsequent homing to dLNs and elicits a broad-spectrum antitumor CD8 T cell response, resulting in significant inhibition of B16 tumor growth. (2) This feasible and efficient co-assembly strategy provides an attractive platform for engineering a range of multivalent vaccines at defined ratios to further enhance antigen-specific T cell responses. This approach may also be used for personalized immunotherapy with neo-epitopes.


Assuntos
Vacinas Anticâncer , Imunoterapia , Neoplasias , Vacinas de Subunidades Antigênicas , Animais , Camundongos , Adjuvantes Imunológicos , Antígenos de Neoplasias , Vacinas Anticâncer/química , Vacinas Anticâncer/uso terapêutico , Linfócitos T CD8-Positivos , Células Dendríticas , Epitopos , Hidrogéis/química , Hidrogéis/uso terapêutico , Imunoterapia/métodos , Camundongos Endogâmicos C57BL , Neoplasias/terapia , Peptídeos/uso terapêutico , Vacinas de Subunidades Antigênicas/química , Vacinas de Subunidades Antigênicas/uso terapêutico
9.
Biometrics ; 79(3): 2664-2676, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-35833513

RESUMO

Cancer (treatment) vaccines that are made of neoantigens, or peptides unique to tumor cells due to somatic mutations, have emerged as a promising method to reinvigorate the immune response against cancer. A key step to prioritizing neoantigens for cancer vaccines is computationally predicting which neoantigens are presented on the cell surface by a human leukocyte antigen (HLA). We propose to address this challenge by training a neural network using mass spectrometry (MS) data composed of peptides presented by at least one of several HLAs of a subject. We embed the neural network within a mixture model and train the neural network by maximizing the likelihood of the mixture model. After evaluating our method using data sets where the peptide presentation status was known, we applied it to analyze somatic mutations of 60 melanoma patients and identified a group of neoantigens more immunogenic in tumor cells than in normal cells. Moreover, neoantigen burden estimated by our method was significantly associated with a measurement of the immune system activity, suggesting these neoantigens could induce an immune response.


Assuntos
Vacinas Anticâncer , Melanoma , Neoplasias , Humanos , Vacinas Anticâncer/química , Antígenos de Neoplasias/genética , Neoplasias/genética , Peptídeos/química , Peptídeos/genética , Antígenos HLA/genética , Melanoma/genética
10.
Iran Biomed J ; 26(2): 160-74, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35090304

RESUMO

Background: Triple-negative breast cancer (TNBC) is determined by the absence of ERBB2, estrogen and progesterone receptors' expression. Cancer vaccines, as the novel immunotherapy strategies, have emerged as promising tools for treating the advanced stage of TNBC. The aim of this study was to evaluate Carcinoembryonic antigen (CEA), Metadherin (MTDH), and Mucin 1 (MUC-1) proteins as vaccine candidates against TNBC. Methods: In this research, a novel vaccine was designed against TNBC by using different immunoinformatics and bioinformatics approaches. Effective immunodominant epitopes were chosen from three antigenic proteins, namely CEA, MTDH, and MUC-1. Recombinant TLR4 agonists were utilized as an adjuvant to stimulate immune responses. Following the selection of antigens and adjuvants, appropriate linkers were chosen to generate the final recombinant protein. To achieve an excellent 3D model, the best predicted 3D model was required to be refined and validated. To demonstrate whether the vaccine/TLR4 complex is stable or not, we performed docking analysis and dynamic molecular simulation. Result: Immunoinformatics and bioinformatics evaluations of the designed construct demonstrated that this vaccine candidate could effectively be used as a therapeutic armament against TNBC. Conclusion: Bioinformatics studies revealed that the designed vaccine has an acceptable quality. Investigating the effectiveness of this vaccine can be confirmed by supplementary in vitro and in vivo studies.


Assuntos
Adjuvantes Imunológicos/química , Vacinas Anticâncer/química , Epitopos/química , Neoplasias de Mama Triplo Negativas/prevenção & controle , Proteínas de Membrana/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Mucina-1/química , Proteínas de Ligação a RNA/química , Receptores de Superfície Celular/química , Desenvolvimento de Vacinas
11.
J Biomol Struct Dyn ; 40(14): 6363-6380, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-33599191

RESUMO

Recently, cancer immunotherapy has gained lots of attention to replace the current chemoradiation approaches and multi-epitope cancer vaccines are manifesting as the next generation of cancer immunotherapy. Therefore, in this study, we used multiple immunoinformatics approaches along with other computational approaches to design a novel multi-epitope vaccine against breast cancer. The most immunogenic regions of the BORIS cancer-testis antigen were selected according to the binding affinity to MHC-I and II molecules as well as containing multiple cytotoxic T lymphocyte (CTL) epitopes by multiple immunoinformatics servers. The selected regions were linked together by GPGPG linker. Also, a T helper epitope (PADRE) and the TLR-4/MD-2 agonist (L7/L12 ribosomal protein from mycobacterium) were incorporated by A(EAAAK)3A linker to form the final vaccine construct. Then, its physicochemical properties, cleavage sites, TAP transport efficiency, B cell epitopes, IFN-γ inducing epitopes and population coverage were predicted. The final vaccine construct was reverse translated, codon-optimized and inserted into pcDNA3.1 to form the DNA vaccine. The final vaccine construct was a stable, immunogenic and non-allergenic protein that contained numerous CTL epitopes, IFN-γ inducing epitopes and several linear and conformational B cell epitopes. Also, the final vaccine construct formed stable and significant interactions with TLR-4/MD-2 complex according to molecular docking and dynamics simulations. Moreover, its world population coverage for HLA-I and HLA-II were about 93% and 96%, respectively. Taking together, these preliminary results can be used as an appropriate platform for further experimental investigations. Communicated by Ramaswamy H. Sarma.


Assuntos
Antígenos de Neoplasias/química , Vacinas Anticâncer/química , Proteínas de Ligação a DNA/química , Neoplasias da Mama/imunologia , Neoplasias da Mama/terapia , Biologia Computacional/métodos , Epitopos de Linfócito B , Epitopos de Linfócito T , Humanos , Simulação de Acoplamento Molecular
12.
Sci Rep ; 11(1): 23121, 2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34848739

RESUMO

In our previous study, immunoinformatic tools were used to design a novel multiepitope cancer vaccine based on the most immunodominant regions of BORIS cancer-testis antigen. The final vaccine construct was an immunogenic, non-allergenic, and stable protein consisted of multiple cytotoxic T lymphocytes epitopes, IFN-γ inducing epitopes, and B cell epitopes according to bioinformatic analyzes. Herein, the DNA sequence of the final vaccine construct was placed into the pcDNA3.1 vector as a DNA vaccine (pcDNA3.1-VAC). Also, the recombinant multiepitope peptide vaccine (MPV) was produced by a transfected BL21 E. coli strain using a recombinant pET-28a vector and then, purified and screened by Fast protein liquid chromatography technique (FPLC) and Western blot, respectively. The anti-tumor effects of prophylactic co-immunization with these DNA and protein cancer vaccines were evaluated in the metastatic non-immunogenic 4T1 mammary carcinoma in BALB/c mice. Co-immunization with the pcDNA3.1-VAC and MPV significantly (P < 0.001) increased the serum levels of the MPV-specific IgG total, IgG2a, and IgG1. The splenocytes of co-immunized mice exhibited a significantly higher efficacy to produce interleukin-4 and interferon-γ and proliferation in response to MPV in comparison with the control. The prophylactic co-immunization regime caused significant breast tumors' growth inhibition, tumors' weight decrease, inhibition of metastasis formation, and enlarging tumor-bearing mice survival time, without any considerable side effects. Taking together, this cancer vaccine can evoke strong immune response against breast tumor and inhibits its growth and metastasis.


Assuntos
Vacinas Anticâncer/imunologia , Proteínas de Ligação a DNA/biossíntese , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/prevenção & controle , Animais , Vacinas Anticâncer/química , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Cromatografia Líquida , Biologia Computacional , Simulação por Computador , Modelos Animais de Doenças , Epitopos , Feminino , Imunidade Humoral , Interferon gama/química , Neoplasias Mamárias Animais/terapia , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/prevenção & controle , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Linfócitos T Citotóxicos/imunologia , Vacinas de Subunidades Antigênicas
13.
Chem Commun (Camb) ; 57(86): 11382-11385, 2021 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-34647563

RESUMO

We report the first synthesis and immunological evaluation of a new glycoconjugate design based on streamlined saponin adjuvants and the Tn carbohydrate antigen. While the novel synthetic constructs induced moderate antibody responses in mice, the versatile chemical platform is amenable to further structure-activity optimizations for the development of self-adjuvanting glycoconjugate cancer vaccines.


Assuntos
Adjuvantes Imunológicos/química , Antígenos Glicosídicos Associados a Tumores/química , Glicoconjugados/química , Saponinas/química , Animais , Formação de Anticorpos , Vacinas Anticâncer/química , Carboidratos/química , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Relação Estrutura-Atividade
14.
ACS Appl Mater Interfaces ; 13(42): 49737-49753, 2021 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-34648269

RESUMO

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.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas Anticâncer/farmacologia , Neoplasias Orofaríngeas/terapia , Papillomaviridae/efeitos dos fármacos , Infecções por Papillomavirus/terapia , Vacinas de Subunidades Antigênicas/farmacologia , Adjuvantes Imunológicos/síntese química , Adjuvantes Imunológicos/química , Animais , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/imunologia , Vacinas Anticâncer/síntese química , Vacinas Anticâncer/química , Linhagem Celular , Humanos , Imunoterapia , Teste de Materiais , Camundongos , Estrutura Molecular , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Neoplasias Orofaríngeas/imunologia , Papillomaviridae/imunologia , Infecções por Papillomavirus/imunologia , Vacinas de Subunidades Antigênicas/síntese química , Vacinas de Subunidades Antigênicas/química
15.
J Mater Chem B ; 9(36): 7435-7446, 2021 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-34551058

RESUMO

Cancer vaccines based on DNA encoding oncogenes have shown great potential in preclinical studies. However, the efficacy of DNA vaccines is limited by their weak immunogenicity because of low cellular internalisation and insufficient activation of dendritic cells (DCs). Calcium phosphate (CP) nanoparticles (NPs) are biodegradable vehicles with low toxicity and high loading capacity of DNA but suffer from stability issues. Here we employed adenosine triphosphate (ATP) as a dual functional agent, i.e. stabiliser for CP and immunological adjuvant, and applied the ATP-modified CP (ACP) NPs to the DNA vaccine. ACP NP-enhanced cellular uptake and improved transfection efficiency of DNA vaccine, and further showed the ability to activate DCs that are critical for them to prime T cells in cancer immunotherapy. As a result, a higher level of antigen-specific antibody with stronger tumour growth inhibition was achieved in mice immunised with the ACP-DNA vaccine. Overall, this one-step synthesised ACP NPs are an efficient nano-delivery system and nano-adjuvant for cancer DNA vaccines.


Assuntos
Trifosfato de Adenosina/química , Adjuvantes Imunológicos/química , Fosfatos de Cálcio/química , Nanopartículas/química , Vacinas de DNA/química , Animais , Reações Antígeno-Anticorpo , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Imunoterapia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/terapia , Transplante Homólogo , Vacinação , Vacinas de DNA/imunologia , Vacinas de DNA/farmacologia
16.
Angew Chem Int Ed Engl ; 60(45): 24179-24188, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34469031

RESUMO

Ganglioside GD2 is an attractive tumor-associated carbohydrate antigen for anti-cancer vaccine development. However, its low immunogenicity and the significant side effects observed with anti-GD2 antibodies present significant obstacles for vaccines. To overcome these, a new GD2 derivative bearing an N-acetamide (NHAc) at its non-reducing end neuraminic acid (9NHAc-GD2) has been designed to mimic the 9-O-acetylated-GD2 (9OAc-GD2), a GD2 based antigen with a restricted expression on tumor cells. 9NHAc-GD2 was synthesized efficiently via a chemoenzymatic method and subsequently conjugated with a powerful carrier bacteriophage Qß. Mouse immunization with the Qß-9NHAc-GD2 conjugate elicited strong and long-lasting IgG antibodies, which were highly selective toward 9NHAc-GD2 with little cross-recognition of GD2. Immunization of canines with Qß-9NHAc-GD2 showed the construct was immunogenic in canines with little adverse effects, paving the way for future clinical translation to humans.


Assuntos
Vacinas Anticâncer/química , Gangliosídeos/síntese química , Vacinas Conjugadas/química , Acetamidas/química , Acetamidas/imunologia , Acetilação , Animais , Vacinas Anticâncer/imunologia , Configuração de Carboidratos , Gangliosídeos/química , Gangliosídeos/imunologia , Hidrólise , Camundongos , Ácidos Neuramínicos/química , Ácidos Neuramínicos/imunologia , Desenvolvimento de Vacinas , Vacinas Conjugadas/imunologia
17.
Int J Biol Sci ; 17(10): 2476-2486, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34326688

RESUMO

Extracellular vesicles (EVs), are membrane-bound vesicles that have many advantages over traditional nanocarriers for drug and gene delivery. Evidence from recent studies indicate that EVs have therapeutic capability with chemical or biological modification. Tumor-derived exosomes (TEXs) were used as a new type of antigens or tumor vaccines in anti-tumor immunotherapy. With superior characteristics, modified EVs were applied to loaded and delivered synthetic drugs, silencing RNA, and microRNA for treatment. Different surface functionalization strategies have been proposed to improve the therapeutic functions of EVs. Appropriately modified EVs for disease intervention provide new avenues for effective clinical treatment strategies. Therefore, this review aimed at elucidating the therapeutic functions of EVs to generate new ideas for treatment and to unlock their hidden potential in translational medicine.


Assuntos
Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Exossomos/química , Vesículas Extracelulares/química , Neoplasias/terapia , Antineoplásicos/química , Vacinas Anticâncer/química , Vacinas Anticâncer/uso terapêutico , Terapia Genética/métodos , Humanos , Ciência Translacional Biomédica
18.
Theranostics ; 11(15): 7425-7438, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34158858

RESUMO

The lack of tumor specific antigens (TSA) and the immune tolerance are two major obstacles for the immunotherapy of cancer. Current immune checkpoint inhibitors (ICIs) show clinical responses in only limited subsets of cancer patients, which, to some extent, depends on the mutation load of tumor cells that may generate neoantigens. Here, we aimed to generate a neoantigen MDP to exhibit stronger anti-tumor efficacy. Methods: In this study, we utilized chemically modified sialic acid precursor tetra acetyl-N-azidoacetyl-mannosamine (AC4ManNAZ) to engineer the glycoproteins on the membranes of tumor cells for the covalent ligation of hapten adjuvant Pam3CSK4 in vivo, which eventually generated a neoantigen, i.e., ManNAZ-DBCO-Pam3CSK4 (MDP), on tumor cells. The high labeling efficiency, relatively specific biodistribution in tumor tissues and the anti-tumor efficacy were confirmed in the syngeneic murine models of the breast cancer and the lung cancer. Results: The generation of MDP neoantigen in tumor-bearing mice significantly evoked both the humoral and the T-cell-dependent antitumor immune responses, resulting in a strong inhibition on the growth of the breast cancer and the lung cancer allografts and significantly prolonged survival of tumor-bearing mice. Interestingly, MDP neoantigen was able to dramatically increase the sensitivity of cancer cells to ICIs and greatly enhance the anti-tumor efficacy in the murine models of both breast cancer and the lung cancer, which showed no or low responses to the immunotherapy with anti-PD1 antibody alone. Conclusions: We developed a simple metabolic glycoengineering method to artificially generate neoantigens on tumor cells to enhance tumor cell immunogenicity, which is able to significantly improve the response and the clinical outcome of ICIs.


Assuntos
Antígenos de Neoplasias , Vacinas Anticâncer , Glicoproteínas , Lipopeptídeos , Neoplasias Mamárias Experimentais , Animais , Antígenos de Neoplasias/química , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/farmacologia , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/farmacologia , Linhagem Celular Tumoral , Feminino , Glicoproteínas/química , Glicoproteínas/imunologia , Glicoproteínas/farmacologia , Lipopeptídeos/química , Lipopeptídeos/imunologia , Lipopeptídeos/farmacologia , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/terapia , Camundongos
19.
Nanomedicine ; 37: 102415, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34174421

RESUMO

Although immune checkpoint inhibitors have significantly improved clinical outcomes in various malignant cancers, only a small proportion of patients reap benefits, likely due to the low number of T cells and high number of immunosuppressive cells in the tumor microenvironment (TME) of patients with advanced disease. We developed a cancer vaccine adjuvanted with nanoemulsion (NE) loaded with TLR7/8 agonist (R848) and analyzed its therapeutic effect alone or in combination with immune checkpoint inhibitors, on antitumor immune responses and the reprogramming of suppressive immune cells in the TME. NE (R848) demonstrated robust local and systemic antitumor immune responses in both subcutaneous and orthotopic mouse lung cancer models, inducing tumor-specific T cell activation and mitigating T cell exhaustion. Combination with anti-PD-1 antibodies showed synergistic effects with respect to therapeutic efficacy and survival rate. Thus, NE (R848)-based cancer vaccines could prevent tumor recurrence and prolong survival by activating antitumor immunity and reprogramming immunosuppression.


Assuntos
Vacinas Anticâncer/farmacologia , Neoplasias Pulmonares/terapia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor 7 Toll-Like/genética , Receptor 8 Toll-Like/genética , Adjuvantes Imunológicos/farmacologia , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Sinergismo Farmacológico , Emulsões/química , Emulsões/farmacologia , Humanos , Imidazóis/farmacologia , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Receptor de Morte Celular Programada 1/imunologia , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Microambiente Tumoral/efeitos dos fármacos
20.
Sci Rep ; 11(1): 12397, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-34117331

RESUMO

Cervical cancer, caused by human papillomavirus (HPV), is the fourth most common type of cancer among women worldwide. While HPV prophylactic vaccines are available, they have no therapeutic effects and do not clear up existing infections. This study aims to design a therapeutic vaccine against cervical cancer using reverse vaccinology. In this study, the E6 and E7 oncoproteins from HPV16 were chosen as the target antigens for epitope prediction. Cytotoxic T lymphocytes (CTL) and helper T lymphocytes (HTL) epitopes were predicted, and the best epitopes were selected based on antigenicity, allergenicity, and toxicity. The final vaccine construct was composed of the selected epitopes, along with the appropriate adjuvant and linkers. The multi-epitope vaccine was evaluated in terms of physicochemical properties, antigenicity, and allergenicity. The tertiary structure of the vaccine construct was predicted. Furthermore, several analyses were also carried out, including molecular docking, molecular dynamics (MD) simulation, and in silico cloning of the vaccine construct. The results showed that the final proposed vaccine could be considered an effective therapeutic vaccine for HPV; however, in vitro and in vivo experiments are required to validate the efficacy of this vaccine candidate.


Assuntos
Vacinas Anticâncer/imunologia , Epitopos/imunologia , Vacinas contra Papillomavirus/imunologia , Neoplasias do Colo do Útero/prevenção & controle , Vacinas Anticâncer/química , Biologia Computacional , Epitopos/química , Feminino , Humanos , Imunogenicidade da Vacina , Simulação de Acoplamento Molecular , Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/imunologia , Proteínas E7 de Papillomavirus/química , Proteínas E7 de Papillomavirus/imunologia , Vacinas contra Papillomavirus/química , Proteínas Repressoras/química , Proteínas Repressoras/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Neoplasias do Colo do Útero/imunologia , Neoplasias do Colo do Útero/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...