Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 311
Filtrar
1.
Immunity ; 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38749447

RESUMEN

Tumors weakly infiltrated by T lymphocytes poorly respond to immunotherapy. We aimed to unveil malignancy-associated programs regulating T cell entrance, arrest, and activation in the tumor environment. Differential expression of cell adhesion and tissue architecture programs, particularly the presence of the membrane tetraspanin claudin (CLDN)18 as a signature gene, demarcated immune-infiltrated from immune-depleted mouse pancreatic tumors. In human pancreatic ductal adenocarcinoma (PDAC) and non-small cell lung cancer, CLDN18 expression positively correlated with more differentiated histology and favorable prognosis. CLDN18 on the cell surface promoted accrual of cytotoxic T lymphocytes (CTLs), facilitating direct CTL contacts with tumor cells by driving the mobilization of the adhesion protein ALCAM to the lipid rafts of the tumor cell membrane through actin. This process favored the formation of robust immunological synapses (ISs) between CTLs and CLDN18-positive cancer cells, resulting in increased T cell activation. Our data reveal an immune role for CLDN18 in orchestrating T cell infiltration and shaping the tumor immune contexture.

2.
Rev Assoc Med Bras (1992) ; 70(3): e20230683, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38655992

RESUMEN

OBJECTIVE: In this study, we aimed to determine the phenolic compounds, the antibacterial activity of extract from Laurus nobilis leaves, and its possible effect on transforming growth factor-ß1 expression level in peripheral blood mononuclear cells. METHODS: The phenolic components of Laurus nobilis were identified by the high-performance liquid chromatography method. The antibacterial activity of this extract was determined by disk diffusion and broth microdilution methods. The transforming growth factor-ß1 expression was analyzed using the RT-qPCR method. RESULTS: Epicatechin was found in the highest amount and o-coumaric acid in the lowest amount. The half-maximal inhibitory concentration (IC50) was determined to be 55.17 µg/mL. The zones of inhibition and minimum inhibitory concentration for Staphylococcus aureus, Enterococcus faecalis, and Klebsiella pneumoniae were 15, 14, and 8 mm and 125, 250, and 1000 µg/mL, respectively. The change in transforming growth factor-ß1 expression levels was found to be statistically significant compared with the control groups (p<0.0001). CONCLUSION: Laurus nobilis extract was found to be effective against bacteria and altered the expression level of transforming growth factor-ß1 in peripheral blood mononuclear cells.


Asunto(s)
Antibacterianos , Enterococcus faecalis , Laurus , Leucocitos Mononucleares , Pruebas de Sensibilidad Microbiana , Extractos Vegetales , Staphylococcus aureus , Factor de Crecimiento Transformador beta1 , Humanos , Antibacterianos/farmacología , Cromatografía Líquida de Alta Presión , Enterococcus faecalis/efectos de los fármacos , Concentración 50 Inhibidora , Klebsiella pneumoniae/efectos de los fármacos , Laurus/química , Leucocitos Mononucleares/efectos de los fármacos , Fenoles/farmacología , Fenoles/análisis , Extractos Vegetales/farmacología , Hojas de la Planta/química , Staphylococcus aureus/efectos de los fármacos , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo
3.
Cancer Cell ; 42(4): 568-582.e11, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38490213

RESUMEN

Major histocompatibility complex (MHC) class I antigen presentation deficiency is a common cancer immune escape mechanism, but the mechanistic implications and potential strategies to address this challenge remain poorly understood. Studying ß2-microglobulin (B2M) deficient mouse tumor models, we find that MHC class I loss leads to a substantial immune desertification of the tumor microenvironment (TME) and broad resistance to immune-, chemo-, and radiotherapy. We show that treatment with long-lasting mRNA-encoded interleukin-2 (IL-2) restores an immune cell infiltrated, IFNγ-promoted, highly proinflammatory TME signature, and when combined with a tumor-targeting monoclonal antibody (mAB), can overcome therapeutic resistance. Unexpectedly, the effectiveness of this treatment is driven by IFNγ-releasing CD8+ T cells that recognize neoantigens cross-presented by TME-resident activated macrophages. These macrophages acquire augmented antigen presentation proficiency and other M1-phenotype-associated features under IL-2 treatment. Our findings highlight the importance of restoring neoantigen-specific immune responses in the treatment of cancers with MHC class I deficiencies.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Animales , Ratones , Antígenos de Histocompatibilidad Clase I/genética , Interleucina-2/genética , Interleucina-2/inmunología , Neoplasias/genética , ARN Mensajero , Microambiente Tumoral
4.
Mol Ther Nucleic Acids ; 35(2): 102162, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38545619

RESUMEN

The co-delivery of microRNAs (miRNAs) and protein-coding RNA presents an opportunity for a combined approach to gene expression and gene regulation for therapeutic applications. Protein delivery is established using long mRNA, self-, and trans-amplifying RNA (taRNA), whereas miRNA delivery typically uses short synthetic oligonucleotides rather than incorporating it as a precursor into long RNA. Although miRNA delivery into the cell cytoplasm using long genomes of RNA viruses has been described, concerns have remained regarding low processing efficiency. However, miRNA precursors can be released from long cytoplasmic alphaviral RNA by a cytoplasmic fraction of Drosha. taRNA, a promising vector platform for infectious disease vaccination, uses a nonreplicating mRNA expressing an alphaviral replicase to amplify a protein-coding short transreplicon-RNA (STR) in trans. To investigate the possibility of simultaneously delivering protein expression and gene silencing, we tested whether a taRNA system can carry and release functional miRNA to target cells. Here, we show that mature miRNA is released from STRs and silences specific targets in a replication-dependent manner for several days without compromising the expression of STR-encoded proteins. Our findings suggest that incorporating miRNAs into the taRNA vector platform has the potential for gene regulation alongside the expression of therapeutic genes.

5.
Cell ; 187(6): 1363-1373.e12, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38366591

RESUMEN

In response to the 2022 outbreak of mpox driven by unprecedented human-to-human monkeypox virus (MPXV) transmission, we designed BNT166, aiming to create a highly immunogenic, safe, accessible, and scalable next-generation vaccine against MPXV and related orthopoxviruses. To address the multiple viral forms and increase the breadth of immune response, two candidate multivalent mRNA vaccines were evaluated pre-clinically: a quadrivalent vaccine (BNT166a; encoding the MPXV antigens A35, B6, M1, H3) and a trivalent vaccine (BNT166c; without H3). Both candidates induced robust T cell responses and IgG antibodies in mice, including neutralizing antibodies to both MPXV and vaccinia virus. In challenge studies, BNT166a and BNT166c provided complete protection from vaccinia, clade I, and clade IIb MPXV. Furthermore, immunization with BNT166a was 100% effective at preventing death and at suppressing lesions in a lethal clade I MPXV challenge in cynomolgus macaques. These findings support the clinical evaluation of BNT166, now underway (NCT05988203).


Asunto(s)
Monkeypox virus , Mpox , Vacuna contra Viruela , Animales , Humanos , Ratones , Macaca fascicularis , Monkeypox virus/genética , Mpox/inmunología , Mpox/prevención & control , Vacunas Combinadas , Virus Vaccinia/genética
7.
SLAS Technol ; 29(1): 100103, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37595636

RESUMEN

Automation of diagnostic assays generally aims to increase reproducibility and throughput while decreasing human errors and hands-on time. Here, we introduce a protocol for the automated chemical conjugation of glycans to color-coded magnetic beads using the KingFisher Flex magnetic particle processor. The resulting glycan-coupled magnetic beads allow the detection of anti-glycan antibodies of different isotypes from various species. By generating anti-glycan antibody profiles, monoclonal antibodies can be screened for their specificity and cross-reactivity, while anti-glycan antibody profiles from different human body fluids can aid in predicting response to treatment or outcome of disease. This efficient, scalable protocol can also be adapted to attach proteins and other biomolecules to beads, making it useful for a wider range of applications that require bead-based laboratory methods.


Asunto(s)
Anticuerpos Monoclonales , Magnetismo , Humanos , Reproducibilidad de los Resultados , Automatización , Polisacáridos/análisis
9.
Front Mol Biosci ; 10: 1291045, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38146535

RESUMEN

Introduction: Exogeneous messenger ribonucleic acid (mRNA) can be used as therapeutic and preventive medication. However, during the enzymatic production process, commonly called in vitro transcription, by-products occur which can reduce the therapeutic efficacy of mRNA. One such by-product is double-stranded RNA (dsRNA). We therefore sought to limit the generation of dsRNA by-products during in vitro transcription. Materials and methods: In vitro transcription was performed with a DNA template including a poly(A)-tail-encoding region, dinucleotide or trinucleotide cap analogs for cotranscriptional capping, and relevant nucleoside triphosphates. Concentrations of UTP or modified UTP (m1ΨTP) and GTP were reduced and fed over the course of the reaction. mRNA was analyzed for dsRNA contamination, yield of the reaction, RNA integrity, and capping efficiency before translational activity was assessed. Results: Limiting the steady-state level of UTP or m1ΨTP during the enzymatic reaction reduced dsRNA formation, while not affecting mRNA yield or RNA integrity. Capping efficiency was optimized with the use of a combined GTP and UTP or m1ΨTP feed, while still reducing dsRNA formation. Lower dsRNA levels led to higher protein expression from the corresponding mRNAs. Discussion: Low steady-state concentrations of UTP and GTP, fed in combination over the course of the in vitro transcription reaction, produce mRNA with high capping and low levels of dsRNA formation, resulting in high levels of protein expression. This novel approach may render laborious purification steps to remove dsRNA unnecessary.

10.
Clin Infect Dis ; 2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-38016021

RESUMEN

BACKGROUND: Protection against contemporary SARS-CoV-2 variants requires sequence-adapted vaccines. METHODS: In this ongoing phase 2/3 trial, 12-17-year-olds (n=108), 18-55-year-olds (n=313), and >55-year-olds (n=306) who previously received 3 original BNT162b2 30-µg doses, received a fourth dose (second booster) of 30-µg bivalent original/Omicron-BA.4/BA.5-adapted BNT162b2 (BNT162b2-Omi.BA.4/BA.5). For comparisons with original BNT162b2, participants were selected from another phase 3 trial. Immunologic superiority 1-month post-vaccination, with respect to 50% neutralizing titers (GMR lower bound [LB] 2-sided 95%CI >1), and noninferiority with respect to seroresponse rates (rate-difference LB 2-sided 95%CI >-5%), for Omicron BA.4/BA.5 were assessed in >55-year-olds versus original BNT162b2 as a second booster. Noninferiority with respect to neutralizing titer level (GMR LB 2-sided 95%CI >0.67) and seroresponse rate (rate-difference LB 2-sided 95%CI >-10%) of Omicron BA.4/BA.5 immune response for BNT162b2-Omi.BA.4/BA.5 in 18‒55-year-olds versus >55-year-olds was assessed. RESULTS: One-month post-vaccination in >55-year-olds, model-adjusted GMR of Omicron BA.4/BA.5 neutralizing titers for the BNT162b2-Omi.BA.4/BA.5 versus BNT162b2 group (2.91; 95%CI 2.45-3.44) demonstrated superiority of BNT162b2-Omi.BA.4/BA.5. Adjusted difference in percentages of >55-year-olds with seroresponse (26.77%; 95%CI 19.59-33.95) showed noninferiority of BNT162b2-Omi.BA.4/BA.5 to BNT162b2. Noninferiority of BNT162b2-Omi.BA.4/BA.5 in 18‒55-year-olds to >55-year-olds was met for model-adjusted GMR and seroresponse. GMTs in 12-17-year-olds increased from baseline to 1-month post-vaccination. The BNT162b2-Omi.BA.4/BA.5 safety profile was similar to booster doses of bivalent Omicron BA.1-modified BNT162b2 and original BNT162b2 reported in previous studies. CONCLUSIONS: Based on immunogenicity and safety data up to 1-month post-vaccination in participants who previously received 3 original BNT162b2 doses, a BNT162b2-Omi.BA.4/BA.5 30 µg booster has a favorable benefit-risk profile. CLINICAL TRIAL REGISTRATION: NCT05472038.

11.
iScience ; 26(11): 108014, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37965155

RESUMEN

Previous studies showed that the neoantigen candidate load is an imperfect predictor of immune checkpoint blockade (ICB) efficacy. Further studies provided evidence that the response to ICB is also affected by the qualitative properties of a few or even single candidates, limiting the predictive power based on candidate quantity alone. Here, we predict ICB efficacy based on neoantigen candidates and their neoantigen features in the context of the mutation type, using Multiple-Instance Learning via Embedded Instance Selection (MILES). Multiple instance learning is a type of supervised machine learning that classifies labeled bags that are formed by a set of unlabeled instances. MILES performed better compared with neoantigen candidate load alone for low-abundant fusion genes in renal cell carcinoma. Our findings suggest that MILES is an appropriate method to predict the efficacy of ICB therapy based on neoantigen candidates without requiring direct T cell response information.

13.
Electromagn Biol Med ; 42(4): 133-143, 2023 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-37811636

RESUMEN

There is strong scientific evidence that the electric field is harmful to life. Exposure to an electric field (EF) can cause lung toxicity and respiratory disorders. In addition, the electric field has been shown to cause tissue damage through inflammation and apoptosis. Juglone (JUG) is one of the powerful antioxidants with anti-apoptotic and anti-inflammatory, various pharmacological properties in the biological system. In this study, we evaluated the efficacy of JUG against the potential adverse effects of electric field on the lung. Twenty-four Wistar albino rats were randomly divided into three groups; control group (Cont), EF group, and EF exposure+JUG-treated group (EJUG). After routine histological procedures, sections stained with hematoxylin-eosin (H&E) showed significant changes in lung tissues in the EF group compared to the Cont group. Significant protective effects were observed in the building volumes and histopathology in the EJUG group. Our immunohistochemical and gene expression results increased the expression of caspase-3 and tumor necrosis factor alpha (TNF-α) in the EF group (p < 0.05). Juglon increased cytokine signal suppressor (SOCS) expression (p < 0.001). These findings were consistent with the antioxidant effect of JUG treatment. We reasoned that exposure to EF damaged rat lung tissues and administration of JUG alleviated the complications caused by 50 Hz EF.


Depending on technological developments, the use of electric devices and equipment is increasing day by day. During the transmission and use of electrical energy, electric and magnetic fields occurs. This forces can affect the environment and people. 50 Hz electric fields damage the cells and tissues of exposed living things in many ways. In our study, we examined the hypothesis that juglone, a natural antioxidant, can reduce the damage caused by the 50 Hz electric field that we are exposed to for a long time in our daily lives. In our study, it was shown by histological and genetic approaches that juglone, which has antioxidant and anti-apoptotic effects, may be protective against lung damage due to electric field exposure on experimental animals. According to our study results, for increased electric field exposure as a consequence of developing technology, juglone could be an important supportive treatment option.


Asunto(s)
Pulmón , Naftoquinonas , Ratas , Animales , Ratas Wistar , Naftoquinonas/farmacología , Electricidad , Antioxidantes/farmacología , Antioxidantes/metabolismo
14.
Mol Ther Nucleic Acids ; 34: 102045, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37876532

RESUMEN

The response to mRNA vaccines needs to be sufficient for immune cell activation and recruitment, but moderate enough to ensure efficacious antigen expression. The choice of the cap structure and use of N1-methylpseudouridine (m1Ψ) instead of uridine, which have been shown to reduce RNA sensing by the cellular innate immune system, has led to improved efficacy of mRNA vaccine platforms. Understanding how RNA modifications influence the cell intrinsic immune response may help in the development of more effective mRNA vaccines. In the current study, we compared mRNA vaccines in mice against influenza virus using three different mRNA formats: uridine-containing mRNA (D1-uRNA), m1Ψ-modified mRNA (D1-modRNA), and D1-modRNA with a cap1 structure (cC1-modRNA). D1-uRNA vaccine induced a significantly different gene expression profile to the modified mRNA vaccines, with an up-regulation of Stat1 and RnaseL, and increased systemic inflammation. This result correlated with significantly reduced antigen-specific antibody responses and reduced protection against influenza virus infection compared with D1-modRNA and cC1-modRNA. Incorporation of m1Ψ alone without cap1 improved antibodies, but both modifications were required for the optimum response. Therefore, the incorporation of m1Ψ and cap1 alters protective immunity from mRNA vaccines by altering the innate immune response to the vaccine material.

15.
Oncoimmunology ; 12(1): 2255041, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37860278

RESUMEN

IMAB362/Zolbetuximab, a first-in-class IgG1 antibody directed against the cancer-associated gastric-lineage marker CLDN18.2, has recently been reported to have met its primary endpoint in two phase 3 trials as a first-line treatment in combination with standard of care chemotherapy in CLDN18.2-positive Her2 negative advanced gastric cancer. Here we characterize the preclinical pharmacology of BNT141, a nucleoside-modified RNA therapeutic encoding the sequence of IMAB362/Zolbetuximab, formulated in lipid nanoparticles (LNP) for liver uptake. We show that the mRNA-encoded antibody displays a stable pharmacokinetic profile in preclinical animal models, mediates CLDN18.2-restricted cytotoxicity comparable to IMAB362 recombinant protein and inhibits human tumor xenograft growth in immunocompromised mice. BNT141 administration did not perpetrate mortality, clinical signs of toxicity, or gastric pathology in animal studies. A phase 1/2 clinical trial with BNT141 mRNA-LNP has been initiated in advanced CLDN18.2-expressing solid cancers (NCT04683939).


Asunto(s)
Neoplasias Gástricas , Animales , Humanos , Ratones , Moléculas de Adhesión Celular , Claudinas/inmunología , ARN Mensajero/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/inmunología , Anticuerpos/genética , Anticuerpos/inmunología
16.
Nat Med ; 29(11): 2844-2853, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37872225

RESUMEN

The oncofetal antigen Claudin 6 (CLDN6) is highly and specifically expressed in many solid tumors, and could be a promising treatment target. We report dose escalation results from the ongoing phase 1/2 BNT211-01 trial evaluating the safety and feasibility of chimeric antigen receptor (CAR) T cells targeting the CLDN6 with or without a CAR-T cell-amplifying RNA vaccine (CARVac) at two dose levels (DLs) in relapsed/refractory CLDN6-positive solid tumors. The primary endpoints were safety and tolerability, maximum tolerated dose and recommended phase 2 dose (RP2D). Secondary endpoints included objective response rate (ORR) and disease control rate. We observed manageable toxicity, with 10 out of 22 patients (46%) experiencing cytokine release syndrome including one grade 3 event and 1 out of 22 (5%) with grade 1 immune effector cell-associated neurotoxicity syndrome. Dose-limiting toxicities occurred in two patients at the higher DL, resolving without sequelae. CAR-T cell engraftment was robust, and the addition of CARVac was well tolerated. The unconfirmed ORR in 21 evaluable patients was 33% (7 of 21), including one complete response. The disease control rate was 67% (14 of 21), with stable disease in seven patients. Patients with germ cell tumors treated at the higher DL exhibited the highest response rate (ORR 57% (4 of 7)). The maximum tolerated dose and RP2D were not established as the trial has been amended to utilize an automated manufacturing process. A repeat of the dose escalation is ongoing and will identify a RP2D for pivotal trials. ClinicalTrials.gov Identifier: NCT04503278 .


Asunto(s)
Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Linfocitos T
17.
Infect Dis Ther ; 12(9): 2241-2258, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37698774

RESUMEN

INTRODUCTION: Vaccination is a critical tool for preventing coronavirus disease 2019 (COVID-19) and influenza illnesses. Coadministration of the COVID-19 vaccine, BNT162b2, with seasonal inactivated influenza vaccine (SIIV) can provide substantial benefits, including streamlining vaccine delivery. METHODS: In this phase 3 study, healthy 18- to 64-year-olds who had received three previous doses of BNT162b2 were randomized (1:1) to the coadministration group (month 0, BNT162b2 + SIIV; month 1, placebo) or the separate-administration group (month 0, placebo + SIIV; month 1, BNT162b2). The primary immunogenicity objective was to demonstrate that the immune responses elicited by BNT162b2 and SIIV [measured by full-length S-binding immunoglobulin G (IgG) levels and strain-specific hemagglutination inhibition assay (HAI) titers against four influenza strains 1 month post-vaccination, respectively] when coadministered were noninferior to those elicited by either vaccine administered alone, based on a prespecified 1.5-fold noninferiority margin [lower bound 95% CI for geometric mean ratio (GMR) > 0.67]. Reactogenicity and adverse event (AE) rates were evaluated. RESULTS: Randomized participants who received study vaccination (N = 1128; coadministration group, n = 564; separate-administration group, n = 564) had a median age of 39 years. Model-adjusted GMRs for coadministration to separate administration were 0.83 (95% CI 0.77, 0.89) for full-length S-binding IgG levels and 0.89-1.00 (lower bound of all 95% CIs > 0.67) for the four influenza strain-specific HAI titers, with all endpoints achieving the prespecified noninferiority criterion. Reactogenicity events were mostly mild or moderate when BNT162b2 was coadministered with SIIV. Serious AEs were reported in < 1% of participants within 1 month after any vaccination; none were considered vaccine-related. CONCLUSIONS: BNT162b2 coadministered with SIIV elicited immune responses that were noninferior to those elicited by BNT162b2 alone and SIIV alone, and BNT162b2 had an acceptable safety profile when coadministered with SIIV. The results of this study support the coadministration of BNT162b2 and SIIV in adults. TRIAL REGISTRATION: ClinicalTrials.gov registration: NCT05310084.

18.
Res Vet Sci ; 164: 104987, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37659348

RESUMEN

Electric field (EF) has been shown to cause tissue damage mainly through oxidative stress, inflammation, and apoptosis. Thus, juglone (5-hydroxy-1,4-naphthoquinone) (JUG), which has antioxidant and antiapoptotic properties, is thought to be effective against electric field-induced damage. We aimed to investigate whether 50 Hz alternating current (AC) triggers inflammation and apoptosis in rat liver and kidney tissues and evaluate the JUG supplement's estimated protective effect. Twenty-four adult male wistar albino rats were divided into control, EF and EF + JUG groups, each containing eight rats. The EF and EF + JUG groups were exposed to EF while no EF exposure and JUG were applied to the control group. At the end of the experiment, liver and kidney tissues were collected for histological (H&E, caspase-3 and TNF-α for immunohistochemical staining), and genetics (SOCS, caspase-3 and TNF-α, PCR analyses). After routine histological procedures, sections stained with H&E showed significant changes in liver and kidney tissues in the EF group compared to the control group (p < 0.05). Significant protective effects were observed in the building volumes and histopathology in the EF + JUG group (p < 0.05). Our gene expression results increased the expression of caspase-3 and TNF-α in the EF group (p < 0.001). Juglone increased SOCS expression (p < 0.001). These findings were consistent with the anti-apoptotic and anti-inflammatory effects of JUG treatment. We reasoned that exposure to EF damaged rat liver and kidney tissues and administration of JUG alleviated the complications caused by 50 Hz EF.


Asunto(s)
Inflamación , Factor de Necrosis Tumoral alfa , Ratas , Masculino , Animales , Caspasa 3/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Riñón , Hígado/metabolismo , Inflamación/metabolismo , Inflamación/veterinaria , Ratas Wistar , Estrés Oxidativo , Apoptosis
19.
Cell Rep ; 42(8): 112888, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37527039

RESUMEN

Evolution of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant has led to the emergence of sublineages with different patterns of neutralizing antibody evasion. We report that Omicron BA.4/BA.5 breakthrough infection of individuals immunized with SARS-CoV-2 wild-type-strain-based mRNA vaccines results in a boost of Omicron BA.4.6, BF.7, BQ.1.1, and BA.2.75 neutralization but does not efficiently boost BA.2.75.2, XBB, or XBB.1.5 neutralization. In silico analyses showed that the Omicron spike glycoprotein lost most neutralizing B cell epitopes, especially in sublineages BA.2.75.2, XBB, and XBB.1.5. In contrast, T cell epitopes are conserved across variants including XBB.1.5. T cell responses of mRNA-vaccinated, SARS-CoV-2-naive individuals against the wild-type strain, Omicron BA.1, and BA.4/BA.5 were comparable, suggesting that T cell immunity against recent sublineages including XBB.1.5 may remain largely unaffected. While some Omicron sublineages effectively evade B cell immunity, spike-protein-specific T cell immunity, due to the nature of polymorphic cell-mediated immune responses, may continue to contribute to prevention/limitation of severe COVID-19 manifestation.


Asunto(s)
COVID-19 , Linfocitos T , Humanos , Glicoproteína de la Espiga del Coronavirus/genética , SARS-CoV-2 , Anticuerpos Neutralizantes , Anticuerpos Antivirales
20.
Pharmaceutics ; 15(8)2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37631282

RESUMEN

Lipid nanoparticles (LNPs) have gained great attention as carriers for mRNA-based therapeutics, finding applications in various indications, extending beyond their recent use in vaccines for infectious diseases. However, many aspects of LNP structure and their effects on efficacy are not well characterized. To further exploit the potential of mRNA therapeutics, better control of the relationship between LNP formulation composition with internal structure and transfection efficiency in vitro is necessary. We compared two well-established ionizable lipids, namely DODMA and MC3, in combination with two helper lipids, DOPE and DOPC, and two polymer-grafted lipids, either with polysarcosine (pSar) or polyethylene glycol (PEG). In addition to standard physicochemical characterization (size, zeta potential, RNA accessibility), small-angle X-ray scattering (SAXS) was used to analyze the structure of the LNPs. To assess biological activity, we performed transfection and cell-binding assays in human peripheral blood mononuclear cells (hPBMCs) using Thy1.1 reporter mRNA and Cy5-labeled mRNA, respectively. With the SAXS measurements, we were able to clearly reveal the effects of substituting the ionizable and helper lipid on the internal structure of the LNPs. In contrast, pSar as stealth moieties affected the LNPs in a different manner, by changing the surface morphology towards higher roughness. pSar LNPs were generally more active, where the highest transfection efficiency was achieved with the LNP formulation composition of MC3/DOPE/pSar. Our study highlights the utility of pSar for improved mRNA LNP products and the importance of pSar as a novel stealth moiety enhancing efficiency in future LNP formulation development. SAXS can provide valuable information for the rational development of such novel formulations by elucidating structural features in different LNP compositions.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...