Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55.649
Filtrar
1.
Biomaterials ; 312: 122742, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39106821

RESUMO

Hypertrophic scar (HS) tends to raised above skin level with high inflammatory microenvironment and excessive proliferation of myofibroblasts. The HS therapy remains challenging due to dense scar tissue which makes it hard to penetrate, and the side effects resulting from intralesional corticosteroid injection which is the mainstay treatment in clinic. Herein, bilayer microneedle patches combined with dexamethasone and colchicine (DC-MNs) with differential dual-release pattern is designed. Two drugs loaded in commercially available materials HA and PLGA, respectively. Specifically, after administration, outer layer rapidly releases the anti-inflammatory drug dexamethasone, which inhibits macrophage polarization to pro-inflammatory phenotype in scar tissue. Subsequently, inner layer degrades sustainedly, releasing antimicrotubular agent colchicine, which suppresses the overproliferation of myofibroblasts with extremely narrow therapeutic window, and inhibits the overexpression of collagen, as well as promotes the regular arrangement of collagen. Only applied once, DC-MNs directly delivered drugs to the scar tissue. Compared to traditional treatment regimen, DC-MNs significantly suppressed HS at lower dosage and frequency by differential dual-release design. Therefore, this study put forward the idea of integrated DC-MNs accompany the development of HS, providing a non-invasive, self-applicable, more efficient and secure strategy for treatment of HS.


Assuntos
Anti-Inflamatórios , Cicatriz Hipertrófica , Colchicina , Dexametasona , Miofibroblastos , Agulhas , Cicatriz Hipertrófica/tratamento farmacológico , Cicatriz Hipertrófica/patologia , Animais , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Dexametasona/farmacologia , Dexametasona/administração & dosagem , Dexametasona/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/farmacologia , Colchicina/farmacologia , Colchicina/administração & dosagem , Camundongos , Sistemas de Liberação de Medicamentos , Humanos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química
2.
Biomaterials ; 312: 122760, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39163825

RESUMO

Inflammation-resident cells within arthritic sites undergo a metabolic shift towards glycolysis, which greatly aggravates rheumatoid arthritis (RA). Reprogramming glucose metabolism can suppress abnormal proliferation and activation of inflammation-related cells without affecting normal cells, holding potential for RA therapy. Single 2-deoxy-d-glucose (2-DG, glycolysis inhibitor) treatment often cause elevated ROS, which is detrimental to RA remission. The rational combination of glycolysis inhibition with anti-inflammatory intervention might cooperatively achieve favorable RA therapy. To improve drug bioavailability and exert synergetic effect, stable co-encapsulation of drugs in long circulation and timely drug release in inflamed milieu is highly desirable. Herein, we designed a stimulus-responsive hyaluronic acid-triglycerol monostearate polymersomes (HTDD) co-delivering 2-DG and dexamethasone (Dex) to arthritic sites. After intravenous injection, HTDD polymersomes facilitated prolonged circulation and preferential distribution in inflamed sites, where overexpressed matrix metalloproteinases and acidic pH triggered drug release. Results indicated 2-DG can inhibit the excessive cell proliferation and activation, and improve Dex bioavailability by reducing Dex efflux. Dex can suppress inflammatory signaling and prevent 2-DG-induced oxidative stress. Thus, the combinational strategy ultimately mitigated RA by inhibiting glycolysis and hindering inflammatory signaling. Our study demonstrated the great potential in RA therapy by reprogramming glucose metabolism in arthritic sites.


Assuntos
Artrite Reumatoide , Desoxiglucose , Dexametasona , Glucose , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/metabolismo , Animais , Glucose/metabolismo , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Camundongos , Desoxiglucose/farmacologia , Inflamação/tratamento farmacológico , Glicólise/efeitos dos fármacos , Polímeros/química , Ácido Hialurônico/química , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Masculino , Humanos , Proliferação de Células/efeitos dos fármacos
3.
PLoS One ; 19(9): e0308069, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39240825

RESUMO

BACKGROUND: Corticosteroids are commonly used to treat COVID-19 patients with hypoxemia, and clinicians have adjusted the corticosteroid intensity on the basis of clinical needs. However, neither the optimal dose nor the duration of treatment has been recommended. OBJECTIVE: To investigate whether cumulative doses of corticosteroids, measured as dexamethasone-equivalent doses over the first 14 days, impact outcomes in patients with COVID-19 pneumonia. METHODS: We conducted a retrospective cohort study of COVID-19 pneumonia patients admitted between April 1st, 2020, and September 30th, 2021. The study focused on the type and dose of corticosteroid administered during the initial 14 days, clinical outcomes, and complications. The primary outcome was in-hospital mortality. RESULTS: Among 271 patients, the mean cumulative dexamethasone-equivalent dose was 158 (119.9-197.25) mg in survivors and 185 (131.7-222.0) mg in nonsurvivors. Univariate analysis revealed that the cumulative dexamethasone-equivalent dose was a risk factor for in-hospital mortality. However, this association did not hold true in the multivariate analysis. After the cumulative dexamethasone-equivalent dose was categorized into quartiles, the moderate dosage (126.01-165.00 mg) in the second quartile was found to be associated with the lowest in-hospital mortality (16.2%). Higher cumulative dexamethasone-equivalent doses were associated with longer hospital and ICU stays and fewer ventilator-free days (p < 0.001). Doses exceeding 165 mg were associated with an increased risk of hospital-acquired infections (p < 0.001). CONCLUSIONS: The cumulative dexamethasone-equivalent dose during the first 14 days is not associated with in-hospital mortality in hypoxemic COVID-19 patients. However, higher cumulative doses exceeding 165 mg are associated with an increased risk of in-hospital mortality and secondary hospital-acquired infections.


Assuntos
Corticosteroides , Tratamento Farmacológico da COVID-19 , COVID-19 , Dexametasona , Mortalidade Hospitalar , Hipóxia , Humanos , Feminino , Masculino , Estudos Retrospectivos , Pessoa de Meia-Idade , Idoso , Dexametasona/administração & dosagem , Dexametasona/uso terapêutico , Dexametasona/efeitos adversos , COVID-19/mortalidade , COVID-19/complicações , Corticosteroides/administração & dosagem , Corticosteroides/efeitos adversos , Corticosteroides/uso terapêutico , Hipóxia/tratamento farmacológico , SARS-CoV-2/isolamento & purificação , Resultado do Tratamento , Relação Dose-Resposta a Droga , Idoso de 80 Anos ou mais
5.
J Hematol Oncol ; 17(1): 79, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39218935

RESUMO

Blinatumomab has emerged as a promising component of first-line therapy for acute B-cell precursor lymphoblastic leukemia (BCP-ALL), bolstering treatment efficacy. To mitigate CD19 selection pressure and reduce the incidence of blinatumomab-associated toxicities, pre-treatment chemotherapy is recommended before administering blinatumomab. From September 2022 to December 2023, we conducted a single-arm, multicenter, phase 2 trial (NCT05557110) in newly diagnosed Philadelphia chromosome-negative BCP-ALL (Ph-negative BCP-ALL) patients. Participants received induction treatment with reduced-dose chemotherapy (RDC), comprising idarubicin, vindesine, and dexamethasone over 7 days, followed by 2 weeks of blinatumomab. Those failing to achieve composite complete remission (CRc) received an additional 2 weeks of blinatumomab. The primary endpoint was the CRc rate post initial induction treatment. Of the 35 enrolled patients, 33 (94%) achieved CRc after 2 weeks of blinatumomab, with 30 (86%) achieving measurable residual disease (MRD) negativity. Two patients extended blinatumomab to 4 weeks. With either 2 or 4 weeks of blinatumomab treatment, all patients achieved CR (35/35) and 89% (31/35) were MRD negativity. The median time to CR was 22 days. Immune effector cell-associated neurotoxicity syndrome was limited (14%, all grade 1). Non-hematological adverse events of grade 3 or higher included pneumonia (17%), sepsis (6%), and cytokine release syndrome (9%). With a median follow-up of 11.5 months, estimated 1-year overall survival and 1-year progression-free survival rates were 97.1% and 82.2%, respectively. These findings affirm that RDC followed by blinatumomab is an effective and well-tolerated induction regimen for newly diagnosed Ph-negative BCP-ALL, supporting a shift towards less intensive and more targeted therapeutic approaches. Trial registration: https://www.clinicaltrials.Gov . Identifier NCT05557110.


Assuntos
Anticorpos Biespecíficos , Protocolos de Quimioterapia Combinada Antineoplásica , Quimioterapia de Indução , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Humanos , Anticorpos Biespecíficos/uso terapêutico , Anticorpos Biespecíficos/administração & dosagem , Anticorpos Biespecíficos/efeitos adversos , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Adulto Jovem , Quimioterapia de Indução/métodos , Idoso , Adolescente , Dexametasona/administração & dosagem , Dexametasona/uso terapêutico , Dexametasona/efeitos adversos , Indução de Remissão
6.
Carbohydr Polym ; 345: 122546, 2024 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-39227091

RESUMO

Herein, dexamethasone (DEX) nanocrystalline suspension (NS)-embedded hydrogel (NS-G) was constructed using a hydroxypropyl methylcellulose (HPMC) polymer to enhance cochlear delivery and attenuate hearing loss following intratympanic (IT) injection. Hydrophobic steroidal nanocrystals were prepared using a bead milling technique and incorporated into a polysaccharide hydrogel. The NS-G system with HPMC (average molecular weight, 86,000 g/mol; 15 mg/mL) was characterized as follows: rod-shaped drug crystalline; particle size <300 nm; and constant complex viscosity ≤1.17 Pa·s. Pulverization of the drug particles into submicron diameters enhanced drug dissolution, while the HPMC matrix increased the residence time in the middle ear cavity, exhibiting a controlled release profile. The IT NS-G system elicited markedly enhanced and prolonged drug delivery (> 9 h) to the cochlear tissue compared with that of DEX sodium phosphate (DEX-SP), a water-soluble prodrug. In mice with kanamycin- and furosemide-induced ototoxicity, NS-G markedly enhanced hearing preservation across all frequencies (8-32 kHz), as revealed by an auditory brainstem response test, compared with both saline and DEX-SP. Moreover, treatment with NS-G showed enhanced anti-inflammatory effects, as evidenced by decreased levels of inflammation-related cytokines. Therefore, the IT administration of DEX NS-loaded HPMC hydrogels is a promising strategy for treating hearing loss.


Assuntos
Cóclea , Dexametasona , Perda Auditiva , Hidrogéis , Derivados da Hipromelose , Injeção Intratimpânica , Nanopartículas , Dexametasona/química , Dexametasona/administração & dosagem , Animais , Derivados da Hipromelose/química , Hidrogéis/química , Nanopartículas/química , Camundongos , Cóclea/efeitos dos fármacos , Cóclea/patologia , Perda Auditiva/tratamento farmacológico , Perda Auditiva/induzido quimicamente , Liberação Controlada de Fármacos , Masculino , Sistemas de Liberação de Medicamentos/métodos
7.
Blood Cancer J ; 14(1): 155, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39261451

RESUMO

Given the early use of triplet and quadruplet regimens, most patients with multiple myeloma (MM) will be exposed and/or refractory to PIs, IMiDs, and anti-CD38 mAbs after first- or second-line treatment. Effective treatment for this group of triple class exposed/refractory (TCE/R) patients is crucial. Here we present a post-hoc subgroup analysis of TCE/R patients treated on the ALGONQUIN study of belantamab mafodotin plus pomalidomide-dexamethasone (belamaf-Pd) for relapsed MM. Of the 99 patients treated on the ALGONQUIN study, 69 were TCE and 56 were TCR and were included in this analysis. Patients had a median of three prior lines of therapy. The ORR was 86.4% in TCE patients and 84.9% in TCR patients, with ≥ very good partial response rates of 64% and 68% respectively. The median progression free survival was 18.3 months in TCE patients and 19.6 months in TCR patients, with overall survival not yet reached and 34.4 months, respectively for TCE and TCR patients. No new safety signals were identified. The most common Grade ≥ 3 AEs were keratopathy (48%), decreased visual acuity (42%), neutropenia (36%), thrombocytopenia (27%), and infection (25%). In this subgroup analysis of the ALGONQUIN study, patients with TCE/TCR disease treated with belamaf-Pd achieved high clinical response rates with durable remissions, comparable to other novel therapeutics in this space.


Assuntos
Anticorpos Monoclonais Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica , Dexametasona , Mieloma Múltiplo , Talidomida , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/mortalidade , Dexametasona/uso terapêutico , Dexametasona/administração & dosagem , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Talidomida/análogos & derivados , Talidomida/uso terapêutico , Talidomida/administração & dosagem , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/administração & dosagem , Adulto , Compostos de Boro/uso terapêutico , Compostos de Boro/administração & dosagem , Idoso de 80 Anos ou mais , Recidiva Local de Neoplasia/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos
8.
Front Immunol ; 15: 1408892, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39234256

RESUMO

Introduction: Ciltacabtagene autoleucel (cilta-cel) is a chimeric antigen receptor T-cell therapy approved for patients with relapsed/refractory multiple myeloma (RRMM). In the phase 3 trial, CARTITUDE-4 (NCT04181827), cilta-cel demonstrated improved efficacy vs. standard of care (SOC; daratumumab plus pomalidomide and dexamethasone [DPd] or pomalidomide plus bortezomib and dexamethasone [PVd]) with a ≥ complete response (≥CR) rate of 73.1% vs. 21.8%. Methods: A cost-per-responder model was developed to assess the value of cilta-cel and SOC (87% DPd and 13% PVd) based on the CARTITUDE-4 trial data from a US mixed payer perspective (76.7% commercial, 23.3% Medicare). The model was developed using progression-free survival (PFS), overall survival (OS), and ≥CR endpoints from CARTITUDE-4 over a period of 25.4 months. Inpatient stays, outpatient visits, drug acquisition, administration, and monitoring costs were included. The base-case model assumed an inpatient setting for each cilta-cel infusion; another scenario included 30% outpatient and 70% inpatient infusions. Costs of managing grade 3-4 adverse events (AEs) and grade 1-4 cytokine release syndrome and neurotoxicity were included. Subsequent therapy costs were incurred after disease progression; terminal care costs were considered upon death events. Outcomes included total cost per treated patient, total cost per complete responder, and cost per month in PFS between cilta-cel and SOC. Costs were adjusted to 2024 US dollars. Results: Total cost per treated patient, total cost per complete responder, and total cost per month in PFS were estimated at $704,641, $963,941, and $30,978 for cilta-cel, respectively, and $840,730, $3,856,559, and $42,520 for SOC over the 25.4-month period. Cost drivers included treatment acquisition costs before progression and subsequent treatment costs ($451,318 and $111,637 for cilta-cel; $529,795 and $265,167 for SOC). A scenario analysis in which 30% of patients received an outpatient infusion (assuming the same payer mix) showed a lower cost per complete responder for cilta-cel ($956,523) than those with an infusion in the inpatient setting exclusively. Discussion: This analysis estimated that cost per treated patient, cost per complete responder, and cost per month in PFS for cilta-cel were remarkably lower than for DPd or PVd, highlighting the substantial clinical and economic benefit of cilta-cel for patients with RRMM.


Assuntos
Análise Custo-Benefício , Imunoterapia Adotiva , Lenalidomida , Mieloma Múltiplo , Talidomida , Humanos , Mieloma Múltiplo/terapia , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/tratamento farmacológico , Lenalidomida/uso terapêutico , Lenalidomida/administração & dosagem , Imunoterapia Adotiva/economia , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Talidomida/análogos & derivados , Talidomida/uso terapêutico , Talidomida/economia , Talidomida/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/economia , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Dexametasona/uso terapêutico , Dexametasona/administração & dosagem , Receptores de Antígenos Quiméricos/uso terapêutico , Receptores de Antígenos Quiméricos/imunologia , Masculino , Feminino , Bortezomib/uso terapêutico , Bortezomib/administração & dosagem , Pessoa de Meia-Idade , Intervalo Livre de Progressão , Resultado do Tratamento , Idoso , Resistencia a Medicamentos Antineoplásicos , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/economia
9.
J Korean Med Sci ; 39(34): e255, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39228186

RESUMO

BACKGROUND: Systemic corticosteroids have become the standard of care for severe to critically ill patients with coronavirus disease 2019 (COVID-19). However, the real-world efficacy and safety outcomes associated with a higher dose of corticosteroids remain uncertain. METHODS: We conducted a nationwide, population-based, matched cohort study of severe to critically ill adult patients with COVID-19 between January 2020 and June 2021 in Korea using the National Health Information Database. Patients using systemic corticosteroids were included and high-dose corticosteroid use was defined as a daily mean prescribed dose of more than 6 mg of dexamethasone. We then employed a proportional hazard regression model to identify prognostic factors for 28-day all-cause mortality and conducted a Fine and Gray regression model to assess risk factors for developing COVID-19-associated pulmonary aspergillosis (CAPA). RESULTS: During the study period, 102,304 patients with COVID-19 were screened, 5,754 met the eligibility criteria, and 2,138 were successfully matched. The mean prescribed daily dose was 4.2 mg and 13.4 mg in the standard- and high-dose groups, respectively, and the mean duration of use was not different between the groups. High-dose corticosteroid use independently increased all-cause mortality at 28 days (adjusted hazard ratio [aHR], 1.48; 95% confidence interval [CI], 1.25-1.76) and 90 days (aHR, 1.63; CI, 1.44-1.85) after admission. Subgroup analysis revealed a statistically significant elevation in the risk of mortality among patients using low-flow or high-flow nasal cannulas, with aHRs of 1.41 and 1.46, respectively. No significant impact of high-dose steroids was observed, even in patients who underwent mechanical ventilation at 28 days (aHR, 1.17; CI, 0.79-1.72). As a safety outcome, high-dose corticosteroid use showed an association with the development of CAPA (aHR, 2.97; 95% CI, 0.94-9.43). CONCLUSION: Among severe to critically ill patients with COVID-19, high-dose corticosteroid use was associated with increased 28-day all-cause mortality and showed a trend toward the development of CAPA.


Assuntos
Corticosteroides , Tratamento Farmacológico da COVID-19 , COVID-19 , Estado Terminal , Dexametasona , SARS-CoV-2 , Humanos , Feminino , Masculino , Pessoa de Meia-Idade , Idoso , COVID-19/mortalidade , COVID-19/complicações , COVID-19/epidemiologia , República da Coreia , Corticosteroides/uso terapêutico , Corticosteroides/administração & dosagem , SARS-CoV-2/isolamento & purificação , Estudos de Coortes , Dexametasona/uso terapêutico , Dexametasona/administração & dosagem , Modelos de Riscos Proporcionais , Adulto , Fatores de Risco
10.
Life Sci ; 356: 123034, 2024 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-39236900

RESUMO

Collagen and hyaluronic acid are essential components of the dermis that collaborate to maintain skin elasticity and hydration due to their unique biochemical properties and interactions within the extracellular matrix. Prolonged exposure to glucocorticoids can induce skin aging, which manifests as diminished collagen content and hyaluronic acid levels in the dermis. Nerol, a monoterpene alcohol found in essential oils, was examined in this study for its potential to counteract glucocorticoid-induced skin aging and the underlying mechanism behind its effects. Our findings reveal that non-toxic concentrations of nerol treatment can reinstate collagen content and hyaluronic acid levels in human dermal fibroblasts treated with dexamethasone. Mechanistically, nerol mitigates dexamethasone-induced oxidative stress by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. The protective effects of nerol were significantly abrogated when the Nrf2 pathway was inhibited using the specific inhibitor ML385. In conclusion, nerol protects human dermal fibroblasts against glucocorticoid-induced skin aging by ameliorating oxidative stress via activation of the Nrf2 pathway, thereby highlighting its potential as a therapeutic agent for preventing and treating glucocorticoid-induced skin aging.


Assuntos
Dexametasona , Fibroblastos , Fator 2 Relacionado a NF-E2 , Estresse Oxidativo , Transdução de Sinais , Envelhecimento da Pele , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Envelhecimento da Pele/efeitos dos fármacos , Dexametasona/farmacologia , Transdução de Sinais/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Glucocorticoides/farmacologia , Pele/efeitos dos fármacos , Pele/metabolismo , Células Cultivadas , Sesquiterpenos/farmacologia , Colágeno/metabolismo
11.
Blood Cancer J ; 14(1): 159, 2024 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-39271684

RESUMO

Lenalidomide, bortezomib, and dexamethasone (RVd) have previously been established as standard-of-care induction therapy for newly diagnosed multiple myeloma (NDMM). More recently, randomized phase 3 data have demonstrated the benefit of the addition of daratumumab (Dara-RVd) to the RVd backbone in terms of improved both depth of response and long-term survival benefit as measured by progression-free survival (PFS). Our group has previously published on a historical cohort of 1000 NDMM patients uniformly treated with RVd induction with impressive both PFS and overall survival. Here, we present a comparative analysis of our RVd cohort with a recent cohort of 326 patients induced with Dara-RVd at our institution with intent to transplant. This analysis demonstrates the utility of this regimen in real-world clinical practice and provides additional insights into D-RVd performance in patient subsets often underrepresented in clinical trials, as well as the impact of daratumumab in maintenance for NDMM patients.


Assuntos
Anticorpos Monoclonais , Protocolos de Quimioterapia Combinada Antineoplásica , Bortezomib , Dexametasona , Lenalidomida , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/mortalidade , Dexametasona/administração & dosagem , Dexametasona/uso terapêutico , Bortezomib/administração & dosagem , Bortezomib/uso terapêutico , Lenalidomida/uso terapêutico , Lenalidomida/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Feminino , Masculino , Pessoa de Meia-Idade , Idoso , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Adulto , Idoso de 80 Anos ou mais , Estudos de Coortes
12.
Int J Mol Sci ; 25(17)2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39273341

RESUMO

Inhalable formulations with cyclodextrins (CDs) as solubility and absorption enhancers show promise for pulmonary delivery. Thiolated hydroxypropyl-ß-cyclodextrin (HP-ß-CD-SH) has mucoadhesive properties, enhancing drug absorption. Moreover, it has self-aggregation capability, which could further improve absorption and drug stability, as well as reduce irritation. This study aims to stabilize CD nanoaggregates using bifunctional cross-linkers and evaluate their benefits for lung drug delivery compared to pristine HP-ß-CD-SH. METHODS: The effectiveness of cross-linked HP-ß-CD-SH nanoparticles (HP-ß-CD-SH-NP) was compared to transient nanoaggregates in enhancing the activity of dexamethasone (DMS) and olive leaf extracts (OLE). DMS, a poorly soluble drug commonly used in lung treatments, and OLE, known for its antioxidant properties, were chosen. Drug-loaded HP-ß-CD-SH-NP were prepared and nebulized onto a lung epithelial Air-Liquid Interface (ALI) model, assessing drug permeation and activity. RESULTS: HP-ß-CD-SH with 25% thiolation was synthesized via microwave reaction, forming 150 nm nanoaggregates and stabilized 400 nm HP-ß-CD-SH-NP. All carriers showed good complexing ability with DMS and OLE and were biocompatible in the lung ALI model. HP-ß-CD-SH promoted DMS absorption, while stabilized HP-ß-CD-SH-NP protected against oxidative stress. CONCLUSION: HP-ß-CD-SH is promising for lung delivery, especially as stabilized nanoaggregates, offering versatile administration for labile molecules like natural extracts.


Assuntos
2-Hidroxipropil-beta-Ciclodextrina , Dexametasona , Sistemas de Liberação de Medicamentos , 2-Hidroxipropil-beta-Ciclodextrina/química , Animais , Humanos , Dexametasona/química , Dexametasona/administração & dosagem , Dexametasona/farmacologia , Dexametasona/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Compostos de Sulfidrila/química , Pulmão/metabolismo , Pulmão/efeitos dos fármacos , Nanopartículas/química , Administração por Inalação , Portadores de Fármacos/química , beta-Ciclodextrinas/química , Ratos
13.
Int Ophthalmol ; 44(1): 377, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39256259

RESUMO

OBJECTIVE: This study seeks to explain the relationship between systemic conditions and hard exudate formations in diabetic macular edema patients. Besides, the study aimed to quantitatively examine changes in the area, location, and impact on visual function of hard exudates following intravitreal dexamethasone implant injections. METHODS: A retrospective analysis was conducted, including 40 patients (40 eyes) diagnosed with non-proliferative diabetic retinopathy and concurrent macular edema between January 1, 2022, and January 1, 2024. Preoperative evaluations included glycated hemoglobin, lipid profile, and renal function examinations. Based on the location of HE, patients were divided into two groups: Group A, with HE in 1 mm of the central fovea, and Group B, with HE outside 1 mm of the central fovea. Selected eyes were subject to pre- and postoperative examinations, including best-corrected visual acuity (BCVA), intraocular pressure, slit-lamp biomicroscopy, scanning laser ophthalmoscopy (SLO), optical coherence tomography, and multifocal electroretinography. Following screening and examination, patients received an immediate intravitreal injection of the DEX implant, with an injection administered at the four-month mark. Hard exudate (HE) areas were measured utilizing SLO fundus imaging. RESULTS: Total cholesterol, low-density lipoprotein, and triglyceride levels were found to be positively correlated with the presence of HE. Following surgical intervention, all patients demonstrated an improvement in BCVA. The mean BCVA increased from a preoperative measurement of 0.79 ± 0.04 to 0.39 ± 0.02 at the 6 month follow-up, indicating a statistically significant difference (p < 0.001). The baseline HE area for the entire patient cohort was 2.28 ± 0.22. One month post-operation, the HE area exhibited a slight increase to 2.27 ± 0.22. However, by the 6 month follow-up, the HE area had significantly decreased to 0.8 ± 0.87, representing a 35.09% reduction from the baseline measurement (p < 0.001). It is worth noting that Patient P1 did not exhibit a statistically significant difference between preoperative and six-month postoperative HE area (p = 0.032). Preoperative BCVA measurements for Group A and Group B were 0.81 ± 0.03 and 0.77 ± 0.03, respectively, with no statistically significant intergroup difference (p = 0.333). The baseline HE area for Group A was 2.61 ± 0.16, which decreased to 0.38 ± 0.20 at the six-month follow-up, representing a 14.60% reduction from the baseline total area. For Group B, the baseline HE area was measured at 1.95 ± 0.09, then decreasing to 1.21 ± 0.13 at the six-month follow-up, indicating a 62.05% reduction from the baseline total area. A statistically significant difference in the postoperative 6 month HE area was observed between Group A and Group B (p < 0.001). In Group A, the reduction in HE area (initial HE area-final HE area) was positively correlated with the improvement in P1 (initial P1-final P1) (r = 0.610, p = 0.004). In Group B, a similar positive correlation was found (initial HE area-final HE area with initial P1-final P1) (r = 0.488, p = 0.029). In Group B, the reduction in HE area (initial HE area-final HE area) correlated positively with the improvement in BCVA (initial BCVA-final BCVA) (r = 0.615, p = 0.004). Additionally, in Group B, the reduction in HE area (initial HE area-final HE area) was positively correlated with the improvement in CMT (initial CMT-final CMT) (r = -0.725, p< 0.001). Aggravated cataracts were observed in thirteen eyes during a follow-up examination 6 months later. CONCLUSION: HE formation is associated with lipid levels. Dexamethasone implants demonstrate effectiveness in reducing HE areas in the short term, reducing macular edema, improving retinal structure, and enhancing visual function. The incidence of postoperative complications such as cataracts and glaucoma remains low.


Assuntos
Dexametasona , Retinopatia Diabética , Implantes de Medicamento , Glucocorticoides , Injeções Intravítreas , Edema Macular , Tomografia de Coerência Óptica , Acuidade Visual , Humanos , Edema Macular/tratamento farmacológico , Edema Macular/etiologia , Edema Macular/diagnóstico , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/diagnóstico , Retinopatia Diabética/complicações , Masculino , Dexametasona/administração & dosagem , Estudos Retrospectivos , Pessoa de Meia-Idade , Feminino , Glucocorticoides/administração & dosagem , Tomografia de Coerência Óptica/métodos , Idoso , Exsudatos e Transudatos , Resultado do Tratamento , Seguimentos
14.
Sci Rep ; 14(1): 20991, 2024 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-39251824

RESUMO

Although COVID-19 infection is an immunosuppressant disease, many immunosuppressant agents, such as pulse methylprednisolone (PMP), dexamethasone (DXM), and tocilizumab (TCZ), were used during the pandemic. Secondary infections in patients with COVID-19 have been reported recently. This study investigated these agents' effects on secondary infections and outcomes in patients with COVID-19 in intensive care units (ICUs). This study was designed retrospectively, and all data were collected from the tertiary intensive care units of six hospitals between March 2020 and October 2021. All patients were divided into three groups: Group I [GI, PMP (-), DXM (-) and TCZ (-)], Group II [GII, PMP (+), DXM (+)], and Group III [GIII, PMP (+), DXM (+), TCZ (+)]. Demographic data, PaO/FiO2 ratio, laboratory parameters, culture results, and outcomes were recorded. To compare GI-GII and GI-GIII, propensity score matching (PSM) was used by matching 14 parameters. Four hundred twelve patients with COVID-19 in the ICU were included in the study. The number of patients with microorganisms ≥ 2 was 279 (67.7%). After PSM, in GII and GIII, the number of (+) tracheal cultures and (+) bloodstream cultures detected different microorganisms ≥ 2 during the ICU period, neuropathy, tracheotomized patients, duration of IMV, and length of ICU stay were significantly higher than GI. The mortality rate was similar in GI and GII, whereas it was significantly higher in GIII than in GI. The use of immunosuppressant agents in COVID-19 patients may lead to an increase in secondary infections. In addition, increased secondary infections may lead to prolonged ICU stay, prolonged IMV duration, and increased mortality.


Assuntos
COVID-19 , Imunossupressores , Unidades de Terapia Intensiva , Humanos , Masculino , Feminino , Estudos Retrospectivos , COVID-19/mortalidade , COVID-19/complicações , COVID-19/epidemiologia , Pessoa de Meia-Idade , Imunossupressores/uso terapêutico , Imunossupressores/efeitos adversos , Idoso , Dexametasona/uso terapêutico , Tratamento Farmacológico da COVID-19 , Metilprednisolona/uso terapêutico , SARS-CoV-2/isolamento & purificação , Anticorpos Monoclonais Humanizados/uso terapêutico , Adulto
15.
BMC Cancer ; 24(1): 1123, 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39251979

RESUMO

Bortezomib, lenalidomide, and dexamethasone (VRD), and bortezomib, doxorubicin, and dexamethasone (PAD), are commonly used in induction regimens for patients with newly diagnosed multiple myeloma (NDMM) in China. This real-world study enrolled 390 patients, 195 receiving VRD and 195 receiving PAD induction. The primary endpoint was progression-free survival (PFS) and stringent complete remission/complete remission. Across the entire cohort, VRD demonstrated significantly improved five-year overall survival (OS) (74% vs. 59%, p = 0.0024) and five-year PFS (67% vs. 37%, p = 0.0018) compared to PAD. Notably, the median OS and PFS were not reached for VRD-treated patients, while they were 77 months (60-not reached [NR]) and 46 months (36-NR), respectively, for PAD. In patients with standard-risk cytogenetics, VRD showed superior five-year OS (83% vs. 58%, p = 0.0038) and PFS (78% vs. 48%, p = 0.0091) compared to PAD. However, these differences were not statistically significant in high-risk patients. For transplanted patients, VRD was associated with superior five-year OS (91% vs. 67%, p = 0.014) and PFS (79% vs. 47%, p = 0.015) compared to PAD. In non-transplanted patients, VRD showed a trend towards improved five-year OS (p = 0.085) and PFS (p = 0.073) compared to the PAD group. In conclusion, VRD displayed superior OS and PFS outcomes in standard-risk patients and those who underwent transplantation. These findings suggest potential advantages of VRD over PAD in real-world clinical settings for NDMM treatment. However, due to the imbalance in transplantation rates between the VRD and PAD groups, limitations in testing for high-risk cytogenetic abnormalities (HRA), and the difference between the received cycles and salvage therapies, the conclusions of this study should be interpreted with caution.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Bortezomib , Dexametasona , Doxorrubicina , Lenalidomida , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/mortalidade , Bortezomib/uso terapêutico , Bortezomib/administração & dosagem , Dexametasona/uso terapêutico , Dexametasona/administração & dosagem , Lenalidomida/uso terapêutico , Lenalidomida/administração & dosagem , Doxorrubicina/uso terapêutico , Doxorrubicina/administração & dosagem , Pessoa de Meia-Idade , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Feminino , Masculino , Idoso , Adulto , Estudos Retrospectivos , Intervalo Livre de Progressão , Idoso de 80 Anos ou mais
16.
Medicine (Baltimore) ; 103(36): e39630, 2024 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-39252248

RESUMO

RATIONALE: Cytokine release syndrome (CRS) is a common adverse event of chimeric antigen receptor T (CAR-T) cell therapy. CRS is generally a systemic inflammatory reaction, but in rare cases, it can occur in specific body areas and is referred to as "local CRS (L-CRS)." A case of laryngeal edema due to L-CRS that required tracheal intubation because of the lack of response to tocilizumab (TCZ) and dexamethasone (DEX) is reported. PATIENT CONCERNS: A 67-year-old woman with relapsed transformed follicular lymphoma was treated with CAR-T cell therapy. Although she had been given TCZ and DEX for CRS, neck swelling appeared on day 4 after infusion. DIAGNOSES: Laryngoscopy showed severe laryngeal edema, which was presumed to be due to L-CRS, since there were no other apparent triggers based on history, physical examination, and computed tomography. INTERVENTIONS: Tracheal intubation was performed because of the risk of upper airway obstruction. Ultimately, 4 doses of tocilizumab (8 mg/kg) and 6 doses of dexamethasone (10 mg/body) were required to improve the L-CRS. OUTCOMES: On day 7, laryngeal edema improved, and the patient could be extubated. LESSONS: The lessons from this case are, first, that CAR-T cell therapy may induce laryngeal edema in L-CRS. Second, TCZ alone may be ineffective in cervical L-CRS. Third, TCZ, as well as DEX, may be inadequate. In such cases, we should recognize L-CRS and manage it early because it may eventually progress to laryngeal edema that requires securing the airway.


Assuntos
Intubação Intratraqueal , Edema Laríngeo , Linfoma Folicular , Humanos , Feminino , Idoso , Edema Laríngeo/etiologia , Edema Laríngeo/terapia , Intubação Intratraqueal/métodos , Intubação Intratraqueal/efeitos adversos , Linfoma Folicular/tratamento farmacológico , Linfoma Folicular/terapia , Linfoma Folicular/complicações , Dexametasona/uso terapêutico , Dexametasona/administração & dosagem , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Síndrome da Liberação de Citocina/etiologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos
17.
Food Res Int ; 194: 114853, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39232503

RESUMO

Cultured fat is an important part of cultured meat, and the ability of adipose-derived mesenchymal stem cells (ADSCs) to differentiate into mature adipose tissue affects the quality of cultured fat. Thus, the primary aim of this study was to screen for combinations of differentiation-inducing factors (DIF) using single-factor experiment and orthogonal experimental design under two-dimensional culture conditions for ADSCs. The results showed that a combination of DIF consisting of 1 µmol/L dexamethasone, 0.1 mmol/L 3-isobutyl-1-methylxanthine, 10 µg/mL insulin, 0.1 mmol/L indomethacin, and 2 µmol/L rosiglitazone was a good choice for the differentiation of ADSCs. An combination of DIF was applied to the preparation of cultured fat with collagen as scaffolds. Forty-eight fatty acids were detected in cultured fat by ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). Among them, the content of twenty-one fatty acids in cultured fat was significantly higher than that of conventional porcine subcutaneous adipose tissue (P < 0.05), and the content of 14 fatty acids was not significantly different (P > 0.05). The ratio of ω-6 polyunsaturated fatty acids content to ω-3 polyunsaturated fatty acids content was 1.23:1, which meant cultured fat was beneficial for human health. This study provides a method to improve the differentiation ability of ADSCs while also providing a reference for indicating the nutritional value of cultured fat.


Assuntos
Diferenciação Celular , Ácidos Graxos , Células-Tronco Mesenquimais , Animais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Suínos , Ácidos Graxos/análise , Células Cultivadas , Tecido Adiposo/citologia , Dexametasona/farmacologia , Espectrometria de Massas em Tandem , Insulina/metabolismo , Rosiglitazona/farmacologia , Indometacina/farmacologia , 1-Metil-3-Isobutilxantina/farmacologia , Cromatografia Líquida de Alta Pressão
18.
Medicine (Baltimore) ; 103(36): e39263, 2024 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-39252252

RESUMO

RATIONALE: Anaphylactic shock, a severe and rapid systemic allergic reaction, poses significant treatment challenges. Epinephrine, the first-line treatment, effectively reverses symptoms but can complicate the clinical picture by elevating lactate levels, blurring the distinction between shock-induced hypoperfusion and drug-induced metabolic effects. PATIENT CONCERNS: A 26-year-old female presented with anaphylactic shock following an antibiotic infusion, experiencing chest tightness, hypotension, and pulmonary edema, without significant past medical history apart from a noted allergy to fish and shrimp. DIAGNOSES: Anaphylaxis was diagnosed based on clinical presentation and supported by imaging that revealed pulmonary edema, despite normal troponin levels and electrocardiogram. INTERVENTIONS: Treatment included 0.5 mg of intramuscular epinephrine and 5 mg of intravenous dexamethasone, with subsequent intubation and mechanical ventilation in the intensive care unit. An intravenous epinephrine infusion was also administered for hemodynamic support. OUTCOMES: While epinephrine resolved the pulmonary edema and stabilized circulation, it led to a significant, albeit transient, increase in lactate levels, which normalized following discontinuation of epinephrine, indicating the metabolic effect of the drug rather than ongoing tissue hypoperfusion. LESSONS: This case illustrates the importance of recognizing epinephrine-induced lactate elevation in anaphylactic shock, necessitating a nuanced interpretation of lactate dynamics. Clinicians must differentiate between lactate elevations due to tissue hypoperfusion and those arising from epinephrine's pharmacologic effects to optimize patient care.


Assuntos
Anafilaxia , Epinefrina , Ácido Láctico , Humanos , Anafilaxia/tratamento farmacológico , Anafilaxia/sangue , Feminino , Adulto , Epinefrina/administração & dosagem , Ácido Láctico/sangue , Dexametasona/uso terapêutico , Dexametasona/administração & dosagem , Edema Pulmonar/induzido quimicamente , Edema Pulmonar/tratamento farmacológico , Antibacterianos/efeitos adversos , Antibacterianos/administração & dosagem
19.
Bone ; 188: 117222, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39102974

RESUMO

BACKGROUND: Zoledronic acid (ZOL) is a type of bisphosphonate with good therapeutic effects on orthopaedic diseases. However, the pharmacological functions of ZOL on steroid-induced avascular necrosis of femoral head (SANFH) and the underlying mechanism remain unclear, which deserve further research. METHODS: SANFH models both in vivo and in vitro were established by dexamethasone (Dex) stimulation. Osteoclastogenesis was examined by TRAP staining. Immunofluorescence was employed to examine autophagy marker (LC3) level. Cell apoptosis was analyzed by TUNEL staining. The interaction between Foxhead box D3 protein (FOXD3) and Annexin A2 (ANXA2) promoter was analyzed using ChIP and dual luciferase reporter gene assays. RESULTS: Dex aggravated osteoclastogenesis and induced osteoclast differentiation and autophagy in vitro, which was abrogated by ZOL treatment. PI3K inhibitor LY294002 abolished the inhibitory effect of ZOL on Dex-induced osteoclast differentiation and autophagy. FOXD3 overexpression neutralized the downregulation effects of ZOL on Dex-induced osteoclasts by transcriptionally activating ANXA2. ANXA2 knockdown reversed the effect of FOXD3 overexpression on ZOL-mediated biological effects in Dex-treated osteoclasts. In addition, ZOL improved SANFH symptoms in rats. CONCLUSION: ZOL alleviated SANFH through regulating FOXD3 mediated ANXA2 transcriptional activity and then promoting PI3K/AKT/mTOR pathway, revealing that FOXD3 might be a target for ZOL in SANFH treatment.


Assuntos
Anexina A2 , Autofagia , Necrose da Cabeça do Fêmur , Fatores de Transcrição Forkhead , Ativação Transcricional , Ácido Zoledrônico , Animais , Necrose da Cabeça do Fêmur/induzido quimicamente , Necrose da Cabeça do Fêmur/patologia , Necrose da Cabeça do Fêmur/genética , Necrose da Cabeça do Fêmur/tratamento farmacológico , Ácido Zoledrônico/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/genética , Autofagia/efeitos dos fármacos , Autofagia/genética , Anexina A2/metabolismo , Anexina A2/genética , Masculino , Ativação Transcricional/efeitos dos fármacos , Dexametasona/farmacologia , Dexametasona/efeitos adversos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Diferenciação Celular/efeitos dos fármacos , Camundongos , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Apoptose/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
20.
BMJ Case Rep ; 17(8)2024 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-39214584

RESUMO

A woman in her mid-60s who is a high hypermetrope presented with bilateral eye pain and headache approximately 1 hour after taking a single dose of a widely available decongestant containing paracetamol, guaifenesin and phenylephrine hydrochloride for coryzal symptoms. She had previous successful bilateral peripheral iridotomies performed for narrow angles. At presentation, her intraocular pressures (IOPs) were significantly raised at 72 mm Hg and 66 mm Hg in the right and left eye, respectively, with bilateral corneal oedema. Her IOP was normalised with urgent treatment using 500 mg intravenous acetazolamide, pilocarpine 2%, dexamethasone 0.1% and IOP-lowering drops. She was listed for cataract surgery and was advised to avoid the precipitating agent and other over-the-counter decongestants. This is the first reported case of bilateral angle closure triggered by a decongestant with such a combination of ingredients. Clinicians should be aware of this rare side effect for prompt diagnosis and management.


Assuntos
Acetaminofen , Acetazolamida , Glaucoma de Ângulo Fechado , Humanos , Glaucoma de Ângulo Fechado/induzido quimicamente , Glaucoma de Ângulo Fechado/tratamento farmacológico , Feminino , Pessoa de Meia-Idade , Acetazolamida/uso terapêutico , Acetazolamida/administração & dosagem , Acetaminofen/efeitos adversos , Acetaminofen/administração & dosagem , Acetaminofen/uso terapêutico , Fenilefrina/efeitos adversos , Fenilefrina/administração & dosagem , Fenilefrina/uso terapêutico , Medicamentos sem Prescrição/efeitos adversos , Medicamentos sem Prescrição/administração & dosagem , Guaifenesina/efeitos adversos , Guaifenesina/administração & dosagem , Guaifenesina/uso terapêutico , Descongestionantes Nasais/efeitos adversos , Descongestionantes Nasais/administração & dosagem , Pressão Intraocular/efeitos dos fármacos , Medicamentos Compostos contra Resfriado, Influenza e Alergia/efeitos adversos , Pilocarpina/uso terapêutico , Pilocarpina/administração & dosagem , Pilocarpina/efeitos adversos , Dexametasona/administração & dosagem , Dexametasona/uso terapêutico , Dexametasona/efeitos adversos , Dor Ocular/induzido quimicamente , Dor Ocular/etiologia , Doença Aguda
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA