Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 508
Filtrar
1.
ACS Nano ; 18(19): 12386-12400, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38699808

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer , Cobre , Macrófagos , Estructuras Metalorgánicas , Piroptosis , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Animales , Ratones , Piroptosis/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Humanos , Cobre/química , Cobre/farmacología , Vacunas contra el Cáncer/química , Microambiente Tumoral/efectos de los fármacos , Nanopartículas/química , Fagocitosis/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias/metabolismo , Ratones Endogámicos BALB C , Eferocitosis , Nanovacunas
2.
Int J Biol Macromol ; 269(Pt 2): 132177, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38729484

RESUMEN

Tumor vaccine, which can effectively prevent tumor recurrence and metastasis, is a promising tool in tumor immunotherapy. However, heterogeneity of tumors and the inability to achieve a cascade effect limit the therapeutic effects of most developing tumor vaccine. We have developed a cascading immunoinducible in-situ mannose-functionalized polydopamine loaded with imiquimod phenylboronic hyaluronic acid nanocomposite gel vaccine (M/P-PDA@IQ PHA) through a boronic ester-based reaction. This reaction utilizes mannose-functionalized polydopamine loaded with imiquimod (M/P-PDA@IQ NAs) as a cross-linking agent to react with phenylboronic-grafted hyaluronic acid. Under near-infrared light irradiation, the M/P-PDA@IQ PHA caused local hyperthermia to trigger immunogenic cell death of tumor cells and tumor-associated antigens (TAAs) releasing. Subsequently, the M/P-PDA@IQ NAs which were gradually released by the pH/ROS/GSH-triggered degradation of M/P-PDA@IQ PHA, could capture and deliver these TAAs to lymph nodes. Finally, the M/P-PDA@IQ NAs facilitated maturation and cross-presentation of dendritic cells, as well as activation of cytotoxic T lymphocytes. Overall, the M/P-PDA@IQ PHA could serve as a novel in situ vaccine to stimulate several key nodes including TAAs release and capture, targeting lymph nodes and enhanced dendritic cells uptake and maturation as well as T cells activation. This cascading immune activation strategy can effectively elicit antitumor immune response.


Asunto(s)
Vacunas contra el Cáncer , Ácido Hialurónico , Hidrogeles , Indoles , Nanopartículas , Polímeros , Ácido Hialurónico/química , Polímeros/química , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Indoles/química , Indoles/farmacología , Animales , Ratones , Hidrogeles/química , Nanopartículas/química , Humanos , Imiquimod/química , Imiquimod/farmacología , Células Dendríticas/inmunología , Vacunación , Línea Celular Tumoral , Inmunoterapia/métodos , Reactivos de Enlaces Cruzados/química , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos
3.
ACS Appl Mater Interfaces ; 16(21): 27187-27201, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38747985

RESUMEN

Development of theranostic nanomedicines to tackle glioma remains to be challenging. Here, we present an advanced blood-brain barrier (BBB)-crossing nanovaccine based on cancer cell membrane-camouflaged poly(N-vinylcaprolactam) (PVCL) nanogels (NGs) incorporated with MnO2 and doxorubicin (DOX). We show that the disulfide bond-cross-linked redox-responsive PVCL NGs can be functionalized with dermorphin and imiquimod R837 through cell membrane functionalization. The formed functionalized PVCL NGs having a size of 220 nm are stable, can deplete glutathione, and responsively release both Mn2+ and DOX under the simulated tumor microenvironment to exert the chemo/chemodynamic therapy mediated by DOX and Mn2+, respectively. The combined therapy induces tumor immunogenic cell death to maturate dendritic cells (DCs) and activate tumor-killing T cells. Further, the nanovaccine composed of cancer cell membranes as tumor antigens, R837 as an adjuvant with abilities of DC maturation and macrophages M1 repolarization, and MnO2 with Mn2+-mediated stimulator of interferon gene activation of tumor cells can effectively act on both targets of tumor cells and immune cells. With the dermorphin-mediated BBB crossing, cell membrane-mediated homologous tumor targeting, and Mn2+-facilitated magnetic resonance (MR) imaging property, the designed NG-based theranostic nanovaccine enables MR imaging and combination chemo-, chemodynamic-, and imnune therapy of orthotopic glioma with a significantly decreased recurrence rate.


Asunto(s)
Glioma , Imagen por Resonancia Magnética , Compuestos de Manganeso , Nanomedicina Teranóstica , Glioma/diagnóstico por imagen , Glioma/tratamiento farmacológico , Glioma/terapia , Glioma/patología , Animales , Ratones , Humanos , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Doxorrubicina/química , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Vacunas contra el Cáncer/química , Inmunoterapia , Óxidos/química , Óxidos/farmacología , Línea Celular Tumoral , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patología , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Barrera Hematoencefálica/metabolismo , Nanogeles/química , Imiquimod/química , Imiquimod/farmacología , Nanovacunas
4.
J Nanobiotechnology ; 22(1): 267, 2024 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-38764014

RESUMEN

Enhancing immune response activation through the synergy of effective antigen delivery and immune enhancement using natural, biodegradable materials with immune-adjuvant capabilities is challenging. Here, we present NAPSL.p that can activate the Toll-like receptor 4 (TLR4) pathway, an amphiphilic exopolysaccharide, as a potential self-assembly adjuvant delivery platform. Its molecular structure and unique properties exhibited remarkable self-assembly, forming a homogeneous nanovaccine with ovalbumin (OVA) as the model antigen. When used as an adjuvant, NAPSL.p significantly increased OVA uptake by dendritic cells. In vivo imaging revealed prolonged pharmacokinetics of NAPSL. p-delivered OVA compared to OVA alone. Notably, NAPSL.p induced elevated levels of specific serum IgG and isotype titers, enhancing rejection of B16-OVA melanoma xenografts in vaccinated mice. Additionally, NAPSL.p formulation improved therapeutic effects, inhibiting tumor growth, and increasing animal survival rates. The nanovaccine elicited CD4+ and CD8+ T cell-based immune responses, demonstrating the potential for melanoma prevention. Furthermore, NAPSL.p-based vaccination showed stronger protective effects against influenza compared to Al (OH)3 adjuvant. Our findings suggest NAPSL.p as a promising, natural self-adjuvanting delivery platform to enhance vaccine design across applications.


Asunto(s)
Adyuvantes Inmunológicos , Melanoma Experimental , Ratones Endogámicos C57BL , Ovalbúmina , Probióticos , Animales , Ovalbúmina/inmunología , Ovalbúmina/química , Ratones , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Probióticos/farmacología , Melanoma Experimental/inmunología , Femenino , Células Dendríticas/inmunología , Receptor Toll-Like 4/metabolismo , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/química , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Humanos , Nanopartículas/química , Linfocitos T CD4-Positivos/inmunología
5.
J Med Chem ; 67(8): 6822-6838, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38588468

RESUMEN

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.


Asunto(s)
Liposomas , Mucina-1 , Animales , Mucina-1/inmunología , Mucina-1/química , Ratones , Humanos , Lipopéptidos/química , Lipopéptidos/inmunología , Lipopéptidos/farmacología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/química , Albúmina Sérica Bovina/química , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Femenino , Ratones Endogámicos BALB C , Antígenos/inmunología , Línea Celular Tumoral
6.
J Am Chem Soc ; 146(17): 11679-11693, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38482849

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer , Lípidos , Liposomas , Nanopartículas , ARN Mensajero , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Nanopartículas/química , Animales , Ratones , ARN Mensajero/genética , ARN Mensajero/inmunología , Lípidos/química , Humanos , Ácido Tióctico/química , Ácido Tióctico/farmacología , Polímeros/química , Guanidinas/química , Guanidinas/farmacología , Línea Celular Tumoral
7.
Adv Mater ; 36(19): e2309927, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38387609

RESUMEN

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.


Asunto(s)
Antígenos de Neoplasias , Inmunoterapia , Interleucina-12 , Animales , Interleucina-12/metabolismo , Ratones , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Humanos , Pliegue de Proteína , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Microambiente Tumoral/efectos de los fármacos , Vacunas contra el Cáncer/química , Nanopartículas/química , Muerte Celular Inmunogénica/efectos de los fármacos , Nanoestructuras/química
8.
Adv Healthc Mater ; 13(13): e2304384, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38301259

RESUMEN

Complete surgical resection of tumor is difficult as the invasiveness of cancer, making the residual tumor a lethal threat to patients. The situation is deteriorated by the immune suppression state after surgery, which further nourishes tumor recurrence and metastasis. Immunotherapy is promising to combat tumor metastasis, but is limited by severe toxicity of traditional immunostimulants and complexity of multiple functional units. Here, it is reported that the simple "trans-surgical bed" delivery of Cu2- xSe nanozyme (CSN) by a microneedle-patch can turn the threat to therapy by efficient in situ vaccination. The biocompatible CSN exhibits both peroxidase and glutathione oxidase-like activities, efficiently exhausting glutathione, boosting free radical generation, and inducing immunogenic cell death. The once-for-all inserting of the patch on surgical bed facilitates sustained catalytic action, leading to drastic decrease of recurrence rate and complete suppression of tumor-rechallenge in cured mice. In vivo mechanism interrogation reveals elevated cytotoxic T cell infiltration, re-educated macrophages, increased dendritic cell maturation, and memory T cells formation. Importantly, preliminary metabolism and safety evaluation validated that the metal accumulation is marginable, and the important biochemical indexes are in normal range during therapy. This study has provided a simple, safe, and robust tumor vaccination approach for postsurgical metastasis control.


Asunto(s)
Vacunas contra el Cáncer , Animales , Ratones , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/química , Cobre/química , Línea Celular Tumoral , Catálisis , Inmunoterapia/métodos , Selenio/química , Selenio/farmacología , Vacunación , Femenino , Ratones Endogámicos C57BL , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/patología , Humanos
9.
Adv Healthc Mater ; 13(14): e2303683, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38386961

RESUMEN

Employing tumor whole cells for tumor immunotherapy is a promising tumor therapy proposed in the early stage, but its therapeutic efficacy is weakened by the methods of eliminating pathogenicity and the mass ratio of the effective antigen carried by itself. Here, by adding gold ion to live cancer cells in the microfluidic droplets, this work obtains dead tumor whole cells with NIR-controlled catalytic ability whose pathogenicity is removed while plenary tumor antigens, major structure, and homing ability are reserved. The engineered tumor cell (Cell-Au) with the addition of prodrug provides 1O2 in an O2-free Russell mechanism, which serves better in a hypoxic tumor microenvironment. This tumor whole-cell catalytic vaccine (TWCV) promotes the activation of dendritic cells and the transformation of macrophages into tumor suppressor phenotype. In 4T1 tumor-bearing mice, the Cell-Au-based vaccine supports the polarization of cytotoxicity T cells, resulting in tumor eradication and long-term animal survival. Compared with antigen vaccines or adoptive cell therapy which takes months to obtain, this TWCV can be prepared in just a few days with satisfactory immune activation and tumor therapeutic efficacy, which provides an alternative way for the preparation of personalized tumor vaccines across tumor types and gives immunotherapy a new path.


Asunto(s)
Vacunas contra el Cáncer , Oro , Inmunoterapia , Animales , Oro/química , Inmunoterapia/métodos , Ratones , Línea Celular Tumoral , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/química , Ratones Endogámicos BALB C , Catálisis , Femenino , Microambiente Tumoral/inmunología , Nanopartículas del Metal/química , Células Dendríticas/inmunología , Humanos , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/patología
10.
Angew Chem Int Ed Engl ; 62(47): e202309744, 2023 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-37781858

RESUMEN

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.


Asunto(s)
Bacteriófagos , Vacunas contra el Cáncer , Neoplasias , Ratones , Humanos , Animales , Antígeno CA-19-9 , Vacunas contra el Cáncer/química , Inmunoglobulina G/metabolismo
11.
Chem Soc Rev ; 52(10): 3353-3396, 2023 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-37070256

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer , Vacunas contra el Cáncer/química , Antígenos de Carbohidratos Asociados a Tumores/química , Vacunas Sintéticas/química , Glicoconjugados/química , Glicósidos
12.
Small Methods ; 7(5): e2201409, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36802205

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Humanos , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/farmacología , Presentación de Antígeno , Adyuvantes Inmunológicos , Antígenos , Neoplasias/terapia , Ovalbúmina/química
13.
Acta Biomater ; 158: 535-546, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36632876

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer , Inmunoterapia , Neoplasias , Vacunas de Subunidad , Animales , Ratones , Adyuvantes Inmunológicos , Antígenos de Neoplasias , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/uso terapéutico , Linfocitos T CD8-positivos , Células Dendríticas , Epítopos , Hidrogeles/química , Hidrogeles/uso terapéutico , Inmunoterapia/métodos , Ratones Endogámicos C57BL , Neoplasias/terapia , Péptidos/uso terapéutico , Vacunas de Subunidad/química , Vacunas de Subunidad/uso terapéutico
14.
Biometrics ; 79(3): 2664-2676, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-35833513

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer , Melanoma , Neoplasias , Humanos , Vacunas contra el Cáncer/química , Antígenos de Neoplasias/genética , Neoplasias/genética , Péptidos/química , Péptidos/genética , Antígenos HLA/genética , Melanoma/genética
15.
Iran Biomed J ; 26(2): 160-74, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35090304

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/química , Vacunas contra el Cáncer/química , Epítopos/química , Neoplasias de la Mama Triple Negativas/prevención & control , Proteínas de la Membrana/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mucina-1/química , Proteínas de Unión al ARN/química , Receptores de Superficie Celular/química , Desarrollo de Vacunas
16.
J Biomol Struct Dyn ; 40(14): 6363-6380, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-33599191

RESUMEN

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.


Asunto(s)
Antígenos de Neoplasias/química , Vacunas contra el Cáncer/química , Proteínas de Unión al ADN/química , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Biología Computacional/métodos , Epítopos de Linfocito B , Epítopos de Linfocito T , Humanos , Simulación del Acoplamiento Molecular
17.
Sci Rep ; 11(1): 23121, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34848739

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Proteínas de Unión al ADN/biosíntesis , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/prevención & control , Animales , Vacunas contra el Cáncer/química , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Cromatografía Liquida , Biología Computacional , Simulación por Computador , Modelos Animales de Enfermedad , Epítopos , Femenino , Inmunidad Humoral , Interferón gamma/química , Neoplasias Mamarias Animales/terapia , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/prevención & control , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Linfocitos T Citotóxicos/inmunología , Vacunas de Subunidad
18.
Chem Commun (Camb) ; 57(86): 11382-11385, 2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34647563

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/química , Antígenos de Carbohidratos Asociados a Tumores/química , Glicoconjugados/química , Saponinas/química , Animales , Formación de Anticuerpos , Vacunas contra el Cáncer/química , Carbohidratos/química , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Relación Estructura-Actividad
19.
ACS Appl Mater Interfaces ; 13(42): 49737-49753, 2021 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-34648269

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas contra el Cáncer/farmacología , Neoplasias Orofaríngeas/terapia , Papillomaviridae/efectos de los fármacos , Infecciones por Papillomavirus/terapia , Vacunas de Subunidad/farmacología , Adyuvantes Inmunológicos/síntesis química , Adyuvantes Inmunológicos/química , Animales , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Vacunas contra el Cáncer/síntesis química , Vacunas contra el Cáncer/química , Línea Celular , Humanos , Inmunoterapia , Ensayo de Materiales , Ratones , Estructura Molecular , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Neoplasias Orofaríngeas/inmunología , Papillomaviridae/inmunología , Infecciones por Papillomavirus/inmunología , Vacunas de Subunidad/síntesis química , Vacunas de Subunidad/química
20.
J Mater Chem B ; 9(36): 7435-7446, 2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34551058

RESUMEN

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.


Asunto(s)
Adenosina Trifosfato/química , Adyuvantes Inmunológicos/química , Fosfatos de Calcio/química , Nanopartículas/química , Vacunas de ADN/química , Animales , Reacciones Antígeno-Anticuerpo , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Inmunoterapia , Ratones , Ratones Endogámicos C57BL , Neoplasias/terapia , Trasplante Homólogo , Vacunación , Vacunas de ADN/inmunología , Vacunas de ADN/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA