Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
BMC Cancer ; 24(1): 1168, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39300392

RESUMO

BACKGROUND: Tri-combination therapy based on hepatic arterial infusion chemotherapy (HAIC) of infusion fluorouracil, leucovorin, and oxaliplatin (FOLFOX-HAIC) plus immune checkpoint inhibitors (ICIs) and tyrosine kinase inhibitors (TKIs) for the locally advanced hepatocellular carcinoma (HCC) patients have been proven effective. However, whether it was best for these HCC patients to start with the most potent therapeutic pattern was still under debate. This retrospective study evaluated the efficacy and safety of FOLFOX-HAIC combined with systemic therapies in the patterns of sequential and concurrent schedules. METHODS: This real-world study included 117 unresectable HCC patients who initially received either FOLFOX-HAIC monotherapy (HAIC group, n = 44) or concurrent ICIs and TKIs (ConHAIC group, n = 73) from March 2020 and June 2022, during the period of FOLFOX-HAIC monotherapy in HAIC group, patients in the HAIC group (n = 30) experienced progressive disease (PD) would have their treatment pattern converted from the FOLFOX-HAIC monotherapy to the combination of FOLFOX-HAIC plus ICIs and TKIs sequentially (SeqHAIC group). The progression-free survival (PFS) and overall survival (OS), as primary outcomes, were compared between patients in the SeqHAIC and ConHAIC groups. RESULTS: The median follow-up time of the SeqHAIC group was 24.92 months (95% CI, 12.74-37.09 months) and of the ConHAIC group was 17.87 months (95% CI, 16.85-18.89 months) and no significant difference was observed in both PFS (HR, 1.572; 95% CI, 0.848-2.916; p = 0.151) and OS (HR, 1.212; 95% CI, 0.574-2.561; p = 0.614) between the SeqHAIC and the ConHAIC groups. As for the tumor responses, there was no significant difference between the two groups regarding tumor responses, overall response rates (p = 0.658) and disease control rates (p = 0.641) were 50.0%, 45.2%, and 83.3%, 89.0% for the SeqHAIC and the ConHAIC groups, respectively. CONCLUSION: Our study revealed that sequential systemic ICIs and TKIs in combination with FOLFOX-HAIC provides similar long-term prognosis and better tolerability compared to concurrent therapy for locally advanced HCC patients. Prospective studies with a larger sample size and longer follow-up are required to validate these findings.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma Hepatocelular , Fluoruracila , Leucovorina , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/mortalidade , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/mortalidade , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Masculino , Feminino , Pessoa de Meia-Idade , Leucovorina/uso terapêutico , Leucovorina/administração & dosagem , Fluoruracila/administração & dosagem , Fluoruracila/uso terapêutico , Estudos Retrospectivos , Idoso , Adulto , Compostos Organoplatínicos/uso terapêutico , Compostos Organoplatínicos/administração & dosagem , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/administração & dosagem , Inibidores de Checkpoint Imunológico/efeitos adversos , Infusões Intra-Arteriais , Oxaliplatina/administração & dosagem , Oxaliplatina/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos
2.
Front Oncol ; 14: 1424824, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39091919

RESUMO

The tumor microenvironment is increasingly acknowledged as a critical contributor to cancer progression, mediating genetic and epigenetic alterations. Beyond diverse cellular interactions from the microenvironment, physicochemical factors such as tumor acidosis also significantly affect cancer dynamics. Recent research has highlighted that tumor acidosis facilitates invasion, immune escape, metastasis, and resistance to therapies. Thus, noninvasive measurement of tumor acidity and the development of targeted interventions represent promising strategies in oncology. Techniques like contrast-enhanced ultrasound (CEUS) can effectively assess blood perfusion, while ultrasound-stimulated microbubble cavitation (USMC) has proven to enhance tumor blood perfusion. We therefore aimed to determine whether CEUS assesses tumor acidity and whether USMC treatment can modulate tumor acidity. Firstly, we tracked CEUS perfusion parameters in MCF7 tumor models and compared them with in vivo tumor pH recorded by pH microsensors. We found that the peak intensity and area under curve of tumor contrast-enhanced ultrasound correlated well with tumor pH. We further conducted USMC treatment on MCF7 tumor-bearing mice, tracked changes of tumor blood perfusion and tumor pH in different perfusion regions before and after the USMC treatment to assess its impact on tumor acidity and optimize therapeutic ultrasound pressure. We discovered that USMC with 1.0 Mpa significantly improved tumor blood perfusion and tumor pH. Furthermore, tumor vascular pathology and PGI2 assays indicated that improved tumor perfusion was mainly due to vasodilation rather than angiogenesis. More importantly, analysis of glycolysis-related metabolites and enzymes demonstrated USMC treatment can reduce tumor acidity by reducing tumor glycolysis. These findings support that CEUS may serve as a potential biomarker to assess tumor acidity and USMC is a promising therapeutic modality for reducing tumor acidosis.

3.
Mikrochim Acta ; 191(7): 387, 2024 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-38869719

RESUMO

A novel construction strategy is introduced for an ultrasensitive dynamic light scattering (DLS) immunosensor targeting alpha fetoprotein (AFP). This approach relies on a self-assembled heptamer fusion protein (A1-C4bpα), incorporating the dual functions of multivalent recognition and crosslinking aggregation amplification due to the presence of seven AFP-specific A1 nanobodies on the A1-C4bpα heptamer. Leveraging antibody-functionalized magnetic nanoparticles for target AFP capture and DLS signal output, the proposed heptamer-assisted DLS immunosensor offers high sensitivity, strong specificity, and ease of operation. Under the optimized conditions, the designed DLS immunosensor demonstrates excellent linear detection of AFP in the concentration range 0.06 ng mL-1 to 512 ng mL-1, with a detection limit of 15 pg mL-1. The selectivity, accuracy, precision, practicability, and reliability of this newly developed method were further validated through an assay of AFP levels in spiked and actual human serum samples. This work introduces a novel approach for constructing ultrasensitive DLS immunosensors, easily extendable to the sensitive determination of other targets via simply replacing the nanobody sequence, holding great promise in various applications, particularly in disease diagnosis.


Assuntos
Difusão Dinâmica da Luz , Limite de Detecção , alfa-Fetoproteínas , alfa-Fetoproteínas/análise , alfa-Fetoproteínas/imunologia , Humanos , Imunoensaio/métodos , Anticorpos Imobilizados/imunologia , Técnicas Biossensoriais/métodos , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/imunologia , Nanopartículas de Magnetita/química
4.
Nano Lett ; 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38838340

RESUMO

Cancer immunotherapies based on cytotoxic CD8+ T lymphocytes (CTLs) are highly promising for cancer treatment. The specific interaction between T-cell receptors and peptide-MHC-I complexes (pMHC-I) on cancer cell membranes critically determines their therapeutic outcomes. However, the lack of appropriate endogenous antigens for MHC-I presentation disables tumor recognition by CTLs. By devising three antigen-loaded self-assembling peptides of pY-K(Ag)-ERGD, pY-K(Ag)-E, and Y-K(Ag)-ERGD to noncovalently generate light-activatable supramolecular antigens at tumor sites in different manners, we report pY-K(Ag)-ERGD as a promising candidate to endow tumor cells with pMHC-I targets on demand. Specifically, pY-K(Ag)-ERGD first generates low-antigenic supramolecular antigens on cancer cell membranes, and a successive light pulse allows antigen payloads to efficiently release from the supramolecular scaffold, directly producing antigenic pMHC-I. Intravenous administration of pY-K(Ag)-ERGD enables light-controlled tumor inhibition when combined with adoptively transferred antigen-specific CTLs. Our strategy is feasible for broadening tumor antigen repertoires for T-cell immunotherapies and advancing precision-controlled T-cell immunotherapies.

5.
J Drug Target ; 32(8): 964-976, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38884143

RESUMO

Numerous nanomedicines have been developed recently that can accumulate selectively in tumours due to the enhanced permeability and retention (EPR) effect. However, the high interstitial fluid pressure (IFP) in solid tumours limits the targeted delivery of nanomedicines. We were previously able to relieve intra-tumoural IFP by low-frequency non-focused ultrasound (LFNFU) through ultrasonic targeted microbubble destruction (UTMD), improving the targeted delivery of FITC-dextran. However, the accumulation of nanoparticles of different sizes and the optimal acoustic pressure were not evaluated. In this study, we synthesised Cy5.5-conjugated mesoporous silica nanoparticles (Cy5.5-MSNs) of different sizes using a one-pot method. The Cy5.5-MSNs exhibited excellent stability and biosafety regardless of size. MCF7 tumour-bearing mice were subjected to UTMD over a range of acoustic pressures (0.5, 0.8, 1.5 and 2.0 MPa), and injected intravenously with Cy5.5-MSNs. Blood perfusion, tumour IFP and intra-tumoural accumulation of Cy5.5-MSNs were analysed. Blood perfusion and IFP initially rose, and then declined, as acoustic pressure intensified. Furthermore, UTMD significantly enhanced the accumulation of differentially sized Cy5.5-MSNs in tumour tissues compared to that of the control group, and the increase was sevenfold higher at an acoustic pressure of 1.5 MPa. Taken together, UTMD enhanced the infiltration and accumulation of Cy5.5-MSNs of different sizes in solid tumours by reducing intra-tumour IFP.


Assuntos
Líquido Extracelular , Microbolhas , Nanopartículas , Dióxido de Silício , Animais , Nanopartículas/química , Camundongos , Humanos , Feminino , Dióxido de Silício/química , Líquido Extracelular/metabolismo , Carbocianinas/química , Carbocianinas/administração & dosagem , Células MCF-7 , Tamanho da Partícula , Sistemas de Liberação de Medicamentos , Pressão , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/patologia , Neoplasias da Mama/patologia , Acústica
6.
Adv Sci (Weinh) ; 11(28): e2401327, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38725147

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal interstitial lung disease, with limited therapeutic options available. Impaired autophagy resulting from aberrant TRB3/p62 protein-protein interactions (PPIs) contributes to the progression of IPF. Restoration of autophagy by modulating the TRB3/p62 PPIs has rarely been reported for the treatment of IPF. Herein, peptide nanofibers are developed that specifically bind to TRB3 protein and explored their potential as a therapeutic approach for IPF. By conjugating with the self-assembling fragment (Ac-GFFY), a TRB3-binding peptide motif A2 allows for the formation of nanofibers with a stable α-helix secondary structure. The resulting peptide (Ac-GFFY-A2) nanofibers exhibit specific high-affinity binding to TRB3 protein in saline buffer and better capacity of cellular uptake to A2 peptide. Furthermore, the TRB3-targeting peptide nanofibers efficiently interfere with the aberrant TRB3/p62 PPIs in activated fibroblasts and fibrotic lung tissue of mice, thereby restoring autophagy dysfunction. The TRB3-targeting peptide nanofibers inhibit myofibroblast differentiation, collagen production, and fibroblast migration in vitro is demonstrated, as well as bleomycin-induced pulmonary fibrosis in vivo. This study provides a supramolecular method to modulate PPIs and highlights a promising strategy for treating IPF diseases by restoring autophagy.


Assuntos
Autofagia , Bleomicina , Modelos Animais de Doenças , Nanofibras , Fibrose Pulmonar , Nanofibras/química , Animais , Autofagia/efeitos dos fármacos , Camundongos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/tratamento farmacológico , Camundongos Endogâmicos C57BL , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/induzido quimicamente , Peptídeos/farmacologia
7.
Neurology ; 102(9): e209299, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38598742

RESUMO

BACKGROUND AND OBJECTIVES: Stroke attributable to nonoptimal temperature needs more attention with dramatic climate change. The aim of this study was to estimate the global burden and distribution characteristics of the burden. METHODS: In this ecological study, we collected data from the Climate Research Unit Gridded Time Series, the World Bank databases, and the Global Burden of Diseases study to estimate the distribution of burden. We used the joinpoint model, decomposition analysis, age-period-cohort model, panel data analysis, and health inequality analysis to assess the different types of stroke burden attributable to different climatic conditions. RESULTS: The burden of stroke attributable to nonoptimal temperature continued to grow, and aging was a key factor in this increase. In 2019, 521,031 (95% uncertainty interval [UI] 402,433-663,996) deaths and 9,423,649 (95% UI 7,207,660-12,055,172) disability-adjusted life years [DALYs] attributable to stroke due to nonoptimal temperature were recorded globally. Globally, men (age-standardized mortality rate [ASMR] 7.70, 95% UI 5.80-9.73; age-standardized DALY rate [ASDR] 139.69, 95% UI 102.96-178.54 in 2019) had a heavier burden than women (ASMR 5.89, 95% UI 4.50-7.60; ASDR 96.02, 95% UI 72.62-123.85 in 2019). Central Asia (ASMR 18.12, 95% UI 13.40-24.53; ASDR 327.35, 95% UI 240.24-440.61 in 2019) had the heaviest burden at the regional level. In the national level, North Macedonia (ASMR 32.97, 95% UI 20.57-47.44 in 2019) and Mongolia (ASDR 568.54, 95% UI 242.03-1,031.14 in 2019) had the highest ASMR/ASDR, respectively. Low temperature currently contributes to the main burden (deaths 474,002, 95% UI 355,077-606,537; DALYs 8,357,198, 95% UI 6,186,217-10,801,911 attributable to low temperature vs deaths 48,030, 95% UI 5,630-104,370; DALYs 1,089,329, 95% UI 112,690-2,375,345 attributable to high temperature in 2019). However, the burden due to high temperature has increased rapidly, especially among people aged older than 10 years, and was disproportionately concentrated in low sociodemographic index (SDI) regions such as Africa. In addition, the rapid increase in the stroke burden due to high temperature in Central Asia also requires special attention. DISCUSSION: This is the first study to assess the global stroke burden attributed to nonoptimal temperature. The dramatic increase in the burden due to high temperature requires special attention, especially in low-SDI countries.


Assuntos
Carga Global da Doença , Acidente Vascular Cerebral , Masculino , Humanos , Feminino , Idoso , Temperatura , Disparidades nos Níveis de Saúde , Anos de Vida Ajustados por Qualidade de Vida , Saúde Global , Acidente Vascular Cerebral/epidemiologia
8.
Adv Mater ; 36(3): e2306736, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37853568

RESUMO

Immune cells are pivotal in cancer immunotherapy, yet their therapeutic effectiveness is often hampered by limited tumor infiltration and inhibitory tumor microenvironments. An alkaline phosphatase (ALP)-responsive and transformable supramolecular bis-specific cell engager (Supra-BiCE) to harness natural killer (NK)/T cells for effective cancer immunotherapy is introduced here. The Supra-BiCE, consisting of both SA-P (a phosphorylated peptide targeting and blocking programmed cell death ligand 1 (PD-L1)) and SA-T (a phosphorylated peptide targeting and blocking T cell immunoglobulin and ITIM domain (TIGIT)) is constructed by a simple co-assembling strategy. Upon intravenous administration, Supra-BiCE self-assembles into nanoribbons and interacts with NK/T cells via TIGIT. Notably, these nanoribbons undergo transformation into long nanofibrils within ALP-overexpressing tumor regions, resulting in enhanced binding affinities of Supra-BiCE to both PD-L1 and TIGIT. Consequently, this leads to the accumulation and retention of NK/T cells within tumor regions. Furthermore, the combinatorial blockade of checkpoints by Supra-BiCE activates infiltrating NK/T cells. Moreover, the adjustable peptide ratio in Supra-BiCE enables customization for optimal therapeutic effects against distinct tumor types. Particularly, Supra-BiCE (T:P = 1:3) achieved 98.27% tumor suppression rate against colon carcinoma model. Overall, this study offers a promising tool for engaging NK and T cells for cancer immunotherapy.


Assuntos
Neoplasias do Colo , Nanotubos de Carbono , Neoplasias , Humanos , Linfócitos T/metabolismo , Células Matadoras Naturais , Antígeno B7-H1 , Imunoterapia/métodos , Receptores Imunológicos/metabolismo , Peptídeos/farmacologia , Microambiente Tumoral
9.
Int Immunopharmacol ; 127: 111413, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38118318

RESUMO

BACKGROUND: The leading course of death in patients with advanced hepatocellular carcinoma (HCC) is intrahepatic progression and associated hepatic failure. The study aimed to evaluate the efficacy of locoregional therapy targeting intrahepatic lesions after intrahepatic progression for advanced HCC. METHODS: Consecutive 263 HCC patients who received lenvatinib combined with immunotherapy were reviewed. Until to last follow-up, 178 patients had disease progression:107 patients had intrahepatic progression (IP group) with or without extrahepatic progression, and 71 patients only had extrahepatic progression (EP group). After intrahepatic progression, 47 patients received systemic therapy (Systemic group), 23 patients received locoregional-systemic therapy (Loco-systemic group), and 37 patients received best supportive therapy (Supportive group). RESULTS: The EP group showed significantly longer OS (overall survival) than the IP group (not reached vs 16.2 months, P = 0.009). Median OS was significantly longer in the Loco-systemic group (20.3 v 8.8 months; P = 0.03) than in the Systemic group. The median PFS (progression-free survival) was 11.7 months in the Loco-systemic group and 5.3 months in the Systemic group (P = 0.046). In patients who progressed fast in first-line treatment, there was no significant difference in OS and PFS between the two groups. CONCLUSION: Intrahepatic progression was associated with a poorer survival outcome compared with extrahepatic progression in advanced HCC. After intrahepatic progression, additional locoregional therapy based on systemic therapy may offer clinical benefits on OS and PFS in second-line treatment, the benefits were limited to patients who had once achieved tumor control during their first-line treatments.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Resultado do Tratamento , Estudos Retrospectivos , Intervalo Livre de Progressão
10.
Cell Death Dis ; 14(12): 794, 2023 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-38049396

RESUMO

Tumor suppression by inducing NCOA4-mediated ferroptosis has been shown to be feasible in a variety of tumors, including gliomas. However, the regulatory mechanism of ferroptosis induced by NCOA4 in glioma has not been studied deeply. HECW1 and ZNF350 are involved in the biological processes of many tumors, but their specific effects and mechanisms on glioma are still unclear. In this study, we found that HECW1 decreased the survival rate of glioma cells and enhanced iron accumulation, lipid peroxidation, whereas ZNF350 showed the opposite effect. Mechanistically, HECW1 directly regulated the ubiquitination and degradation of ZNF350, eliminated the transcriptional inhibition of NCOA4 by ZNF350, and ultimately activated NCOA4-mediated iron accumulation, lipid peroxidation, and ferroptosis. We demonstrate that HECW1 induces ferroptosis and highlight the value of HECW1 and ZNF350 in the prognostic evaluation of patients with glioma. We also elucidate the mechanisms underlying the HECW1/ZNF350/NCOA4 axis and its regulation of ferroptosis. Our findings enrich the understanding of ferroptosis and provide potential treatment options for glioma patients.


Assuntos
Ferroptose , Glioma , Proteínas do Tecido Nervoso , Coativadores de Receptor Nuclear , Proteínas Repressoras , Ubiquitina-Proteína Ligases , Humanos , Ferroptose/genética , Glioma/patologia , Ferro/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Coativadores de Receptor Nuclear/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Proteínas Repressoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA