Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 13.877
Filtrar
1.
Int J Nanomedicine ; 19: 9889-9919, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39351000

RESUMEN

With the in-depth knowledge of the pathological and physiological characteristics of the intestinal barrier-portal vein/intestinal lymphatic vessels-systemic circulation axis, oral targeted drug delivery is frequently being renewed. With many advantages, such as high safety, convenient administration, and good patient compliance, many researchers have begun to explore targeted drug delivery from intravenous injections to oral administration. Over the past few decades, the fields of materials science and nanomedicine have produced various drug delivery platforms that hold great potential in overcoming the multiple barriers associated with oral drug delivery. However, the oral transport of particles into the systemic circulation is extremely difficult due to immune rejection and biochemical invasion in the intestine, which limits absorption and entry into the bloodstream. The feasibility of the oral delivery of targeted drugs to sites outside the gastrointestinal tract (GIT) is unknown. This article reviews the biological barriers to drug absorption, the in vivo fate and transport mechanisms of drug carriers, the theoretical basis for oral administration, and the impact of carrier structural evolution on oral administration to achieve this goal. Finally, this article reviews the characteristics of different nano-delivery systems that can enhance the bioavailability of oral therapeutics and highlights their applications in the efficient creation of oral anticancer nanomedicines.


Asunto(s)
Antineoplásicos , Nanomedicina , Neoplasias , Humanos , Nanomedicina/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos/química , Administración Oral , Animales , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos/métodos , Disponibilidad Biológica , Nanopartículas/química , Nanopartículas/administración & dosificación
2.
AAPS PharmSciTech ; 25(7): 233, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39358486

RESUMEN

Docetaxel (DTX) has become widely accepted as a first-line treatment for metastatic breast cancer; however, the frequent development of resistance provides challenges in treating the disease.C60 fullerene introduces a unique molecular form of carbon, exhibiting attractive chemical and physical properties. Our study aimed to develop dicarboxylic acid-derivatized C60 fullerenes as a novel DTX delivery carrier. This study investigated the potential of water-soluble fullerenes to deliver the anti-cancer drug DTX through a hydrophilic linker. The synthesis was carried out using the Prato reaction. The spectroscopic analysis confirmed the successful conjugation of DTX molecules over fullerenes. The particle size of nanoconjugate was reported to be 122.13 ± 1.63 nm with a conjugation efficiency of 76.7 ± 0.14%. The designed conjugate offers pH-dependent release with significantly less plasma pH, ensuring maximum release at the target site. In-vitro cell viability studies demonstrated the enhanced cytotoxic nature of the developed nanoconjugate compared to DTX. These synthesized nanoscaffolds were highly compatible with erythrocytes, indicating the safer intravenous route administration. Pharmacokinetic studies confirmed the higher bioavailability (~ 6 times) and decreased drug clearance from the system vis-à-vis plain drug. The histological studies reveal that nanoconjugate-treated tumour cells exhibit similar morphology to normal cells. Therefore, it was concluded that this developed formulation would be a valuable option for clinical use.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Ácidos Carboxílicos , Supervivencia Celular , Docetaxel , Sistemas de Liberación de Medicamentos , Fulerenos , Fulerenos/química , Fulerenos/administración & dosificación , Docetaxel/administración & dosificación , Docetaxel/farmacocinética , Docetaxel/farmacología , Docetaxel/química , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Humanos , Femenino , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Antineoplásicos/química , Animales , Supervivencia Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Ácidos Carboxílicos/química , Tamaño de la Partícula , Portadores de Fármacos/química , Línea Celular Tumoral , Liberación de Fármacos , Nanoconjugados/química , Ratas , Células MCF-7 , Disponibilidad Biológica
3.
Sci Rep ; 14(1): 22048, 2024 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-39333571

RESUMEN

Nanoparticle-mediated drug delivery offers a promising approach to targeted cancer therapy, leveraging the ability of nanoparticles to deliver therapeutic agents directly to cancerous tissues with minimal impact on surrounding healthy cells. The presence of these nanoparticles is governed by a concentration equation, which accounts for the diffusion, convection, and reaction of the nanoparticles with the blood components. It is well-known that whenever a disease or infection occurs in a human, in 80% of cases a rise in the concentration of hydrogen peroxide in the blood occurs. This is the reason why blood is assumed to contain hydrogen peroxide (in the present study), which is a biomarker of oxidative stress and inflammation. This study explores the integration of machine learning (ML) techniques into the optimization of drug delivery processes within the human cardiovascular system, focusing on the enhancement of these processes through the application of magnetic fields. By employing ML algorithms, we analyze and predict the behavior of nanoparticles as they navigate the complex fluid dynamics of the cardiovascular system, particularly under the influence of an external magnetic field. The predictive power of ML models enables the precise control of nanoparticle trajectories, optimizing their accumulation in cancerous tissues and improving the efficacy of the drug delivery system. The findings of this study demonstrate that ML-enhanced magnetic targeting can significantly enhance the precision and effectiveness of nanoparticle-mediated drug delivery, offering a new paradigm in cancer treatment strategies. This approach has the potential to revolutionize the field by providing personalized and highly efficient therapeutic solutions for cancer patients.


Asunto(s)
Sistemas de Liberación de Medicamentos , Aprendizaje Automático , Campos Magnéticos , Nanopartículas , Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Nanopartículas/química , Sistemas de Liberación de Medicamentos/métodos , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/efectos de los fármacos , Sistema de Administración de Fármacos con Nanopartículas , Peróxido de Hidrógeno/metabolismo , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética
4.
Int J Nanomedicine ; 19: 9821-9841, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39345910

RESUMEN

Purpose: Renal cell carcinoma (RCC) is the most common and lethal type of urogenital cancer, with one-third of new cases presenting as metastatic RCC (mRCC), which, being the seventh most common cancer in men and the ninth in women, poses a significant challenge. For patients with poor prognosis, temsirolimus (TEM) has been approved for first-line therapy, possessing pharmacodynamic activities that block cancer cell growth and inhibit proliferation-associated proteins. However, TEM suffers from poor water solubility, low bioavailability, and systemic side effects. This study aims to develop a novel drug formulation for the treatment of RCC. Methods: In this study, amphiphilic block copolymer (poly(ethylene glycol) monomethyl ether-poly(beta-amino ester)) (mPEG-PBAE) was utilized as a drug delivery vehicle and TEM-loaded micelles were prepared by thin-film hydration method by loading TEM inside the nanoparticles. Then, the molecular weight of mPEG-PBAE was controlled to make it realize hydrophobic-hydrophilic transition in the corresponding pH range thereby constructing pH-responsive TEM-loaded micelles. Characterization of pH-responsive TEM-loaded nanomicelles particle size, potential and micromorphology while its determination of drug-loading properties, in vitro release properties. Finally, pharmacodynamics and hepatorenal toxicity were further evaluated. Results: TEM loading in mPEG-PBAE increased the solubility of TEM in water from 2.6 µg/mL to more than 5 mg/mL. The pH-responsive TEM-loaded nanomicelles were in the form of spheres or spheroidal shapes with an average particle size of 43.83 nm and a Zeta potential of 1.79 mV. The entrapment efficiency (EE) of pH-responsive TEM nanomicelles with 12.5% drug loading reached 95.27%. Under the environment of pH 6.7, the TEM was released rapidly within 12 h, and the release rate could reach 73.12% with significant pH-dependent characteristics. In vitro experiments showed that mPEG-PBAE preparation of TEM-loaded micelles had non-hemolytic properties and had significant inhibitory effects on cancer cells. In vivo experiments demonstrated that pH-responsive TEM-loaded micelles had excellent antitumor effects with significantly reduced liver and kidney toxicity. Conclusion: In conclusion, we successfully prepared pH-responsive TEM-loaded micelles. The results showed that pH-responsive TEM-loaded micelles can achieve passive tumor targeting of TEM, and take advantage of the acidic conditions in tumor tissues to achieve rapid drug release.


Asunto(s)
Antineoplásicos , Carcinoma de Células Renales , Neoplasias Renales , Micelas , Polietilenglicoles , Sirolimus , Sirolimus/administración & dosificación , Sirolimus/química , Sirolimus/farmacocinética , Sirolimus/farmacología , Sirolimus/análogos & derivados , Humanos , Polietilenglicoles/química , Concentración de Iones de Hidrógeno , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Antineoplásicos/administración & dosificación , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Carcinoma de Células Renales/tratamiento farmacológico , Línea Celular Tumoral , Tamaño de la Partícula , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Ratones , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Femenino , Masculino
5.
BMC Cancer ; 24(1): 1131, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261851

RESUMEN

BACKGROUND: Sunitinib, a newly developed multi-targeted tyrosine kinase inhibitor (TKI), has become a common therapeutic option for managing advanced renal cell carcinoma (RCC). Examining the mechanism underlying the interaction between sunitinib and isavuconazole was the aim of this effort. METHODS: The concentrations of sunitinib and its primary metabolite, N-desethyl sunitinib, were analyzed and quantified using ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). Our study evaluated the potential interaction between isavuconazole and sunitinib using rat liver microsomes (RLM), human liver microsomes (HLM), and in vivo rat models. For the in vivo study, two groups (n = 5) of Sprague-Dawley (SD) rats were randomly allocated to receive sunitinib either with or without co-administration of isavuconazole. Additionally, the effects of isavuconazole on the metabolic stability of sunitinib and N-desethyl sunitinib were studied in RLM in vitro. RESULTS: Our findings demonstrated that in RLM, isavuconazole exhibited a mixed non-competitive and competitive inhibition mechanism, with an IC50 (half maximal inhibitory concentration) value of 1.33 µM. Meanwhile, in HLM, isavuconazole demonstrated a competitive inhibition mechanism, with an IC50 of 5.30 µM. In vivo studies showed that the presence of isavuconazole significantly increased the pharmacokinetic characteristics of sunitinib, with the AUC(0→t), AUC(0→∞), and Tmax rising to approximately 211.38%, 203.92%, and 288.89%, respectively, in contrast to the control group (5 mg/kg sunitinib alone). The pharmacokinetic characteristics of the metabolite N-desethyl sunitinib in the presence of isavuconazole remained largely unchanged compared to the control group. Furthermore, in vitro metabolic stability experiments revealed that isavuconazole inhibited the metabolic processing of both sunitinib and N-desethyl sunitinib. CONCLUSIONS: Isavuconazole had a major impact on sunitinib metabolism, providing fundamental information for the precise therapeutic administration of sunitinib.


Asunto(s)
Interacciones Farmacológicas , Indoles , Microsomas Hepáticos , Nitrilos , Piridinas , Pirroles , Sunitinib , Triazoles , Sunitinib/farmacología , Sunitinib/farmacocinética , Animales , Piridinas/farmacocinética , Piridinas/farmacología , Ratas , Nitrilos/farmacocinética , Nitrilos/farmacología , Humanos , Microsomas Hepáticos/metabolismo , Microsomas Hepáticos/efectos de los fármacos , Pirroles/farmacocinética , Pirroles/farmacología , Triazoles/farmacocinética , Triazoles/farmacología , Indoles/farmacocinética , Indoles/farmacología , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Masculino , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo
6.
Nat Commun ; 15(1): 8172, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39289401

RESUMEN

Nanocarriers (NCs) play a crucial role in delivering theranostic agents to tumors, making them a pivotal focus of research. However, the persistently low delivery efficiency of engineered NCs has been a significant challenge in the advancement of nanomedicine, stirring considerable debate. Transvascular transport is a critical pathway for NC delivery from vessels to tumors, yet a comprehensive understanding of the interactions between NCs and vascular systems remains elusive. In recent years, considerable efforts have been invested in elucidating the transvascular transport mechanisms of NCs, leading to promising advancements in tumor delivery and theranostics. In this context, we highlight various delivery mechanisms, including the enhanced permeability and retention effect, cooperative immune-driven effect, active transcytosis, and cell/bacteria-mediated delivery. Furthermore, we explore corresponding strategies aimed at enhancing transvascular transport of NCs for efficient tumor delivery. These approaches offer intriguing solutions spanning physicochemical, biological, and pharmacological domains to improve delivery and therapeutic outcomes. Additionally, we propose a forward-looking delivery framework that relies on advanced tumor/vessel models, high-throughput NC libraries, nano-bio interaction datasets, and artificial intelligence, which aims to guide the design of next-generation carriers and implementation strategies for optimized delivery.


Asunto(s)
Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Nanopartículas , Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Nanopartículas/química , Portadores de Fármacos/química , Animales , Sistemas de Liberación de Medicamentos/métodos , Transcitosis , Transporte Biológico , Nanomedicina Teranóstica/métodos , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Nanomedicina/métodos
7.
Int J Nanomedicine ; 19: 9613-9635, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39309184

RESUMEN

Background: The combination of nanoplatform-based chemotherapy and photodynamic therapy (PDT) is a promising way to treat cancer. Celastrol (Cela) exhibits highly effective anti-hepatoma activity with low water solubility, poor bioavailability, non-tumor targeting, and toxic side effects. The combination of Cela-based chemotherapy and PDT via hepatoma-targeting and reactive oxygen species (ROS)-responsive polymeric micelles (PMs) could solve the application problem of Cela and further enhance antitumor efficacy. Methods: In this study, Cela and photosensitizer chlorin e6 (Ce6) co-loaded glycyrrhetinic acid-modified carboxymethyl chitosan-thioketal-rhein (GCTR) PMs (Cela/Ce6/GCTR PMs) were prepared and characterized. The safety, ROS-sensitive drug release, and intracellular ROS production were evaluated. Furthermore, the in vitro anti-hepatoma effect and cellular uptaken in HepG2 and BEL-7402 cells, and in vivo pharmacokinetic, tissue distribution, and antitumor efficacy of Cela/Ce6/GCTR PMs in H22 tumor-bearing mice were then investigated. Results: Cela/Ce6/GCTR PMs were successfully prepared with nanometer-scale particle size, favorable drug loading capacity, and encapsulation efficiency. Cela/Ce6/GCTR PMs exhibited a strong safety profile and better hemocompatibility, exhibiting less damage to normal tissues. Compared with Cela-loaded GCTR PMs, the ROS-responsiveness of Cela/Ce6/GCTR PMs was increased, and the release of Cela was accelerated after combination with PDT. Cela/Ce6/GCTR PMs can efficiently target liver tumor cells by uptake and have a high cell-killing effect in response to ROS. The combination of GCTR PM-based chemotherapy and PDT resulted in increased bioavailability of Cela and Ce6, improved liver tumor targeting, and better anti-hepatoma effects in vivo. Conclusion: Hepatoma-targeting and ROS-responsive GCTR PMs co-loaded with Cela and Ce6 combined with PDT exhibited improved primary hepatic carcinoma therapeutic effects with lower toxicity to normal tissues, overcoming the limitations of monotherapy and providing new strategies for tumor treatment.


Asunto(s)
Carcinoma Hepatocelular , Quitosano , Clorofilidas , Neoplasias Hepáticas , Micelas , Fotoquimioterapia , Fármacos Fotosensibilizantes , Porfirinas , Especies Reactivas de Oxígeno , Animales , Fotoquimioterapia/métodos , Especies Reactivas de Oxígeno/metabolismo , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Ratones , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacocinética , Fármacos Fotosensibilizantes/administración & dosificación , Células Hep G2 , Porfirinas/química , Porfirinas/farmacocinética , Porfirinas/farmacología , Porfirinas/administración & dosificación , Quitosano/química , Quitosano/análogos & derivados , Línea Celular Tumoral , Triterpenos Pentacíclicos/farmacología , Triterpenos Pentacíclicos/farmacocinética , Triterpenos/química , Triterpenos/farmacología , Triterpenos/farmacocinética , Ácido Glicirretínico/química , Ácido Glicirretínico/farmacología , Ácido Glicirretínico/farmacocinética , Ácido Glicirretínico/análogos & derivados , Polímeros/química , Distribución Tisular , Liberación de Fármacos , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Masculino , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética
8.
Clin Transl Sci ; 17(9): e70006, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39286959

RESUMEN

Venetoclax, a small molecule inhibitor of BCL-2, has demonstrated efficacy in treating acute leukemias and has been recommended as one of the first-line anti-leukemia therapies. Although venetoclax has been suggested to probably possess the ability to penetrate the central nervous system (CNS), current data to elucidate the characteristics of venetoclax in cerebrospinal fluid (CSF), bone marrow (BM), and plasma are still lacking. This study investigated the real-world characteristics of venetoclax concentrations in CSF, BM, and plasma in acute leukemia patients. Thirteen acute leukemia patients treated with venetoclax were included, with paired samples of CSF, BM, and plasma collected and venetoclax concentrations measured using LC-MS/MS. With the results, the median venetoclax concentrations were 2030 ng/mL in plasma, 16.7 ng/mL in CSF, and 1390 ng/mL in BM. The percentages of CSF/plasma and BM/plasma were 0.74% and 70.37%, respectively. While no direct correlation was observed between CSF and plasma venetoclax levels, there was a trend toward an improved CSF/plasma percentage over time following the last administration of venetoclax. In contrast, a strong correlation was found between BM and plasma levels. This study demonstrated that venetoclax could reach its effective concentration in most patients, suggesting its potential clinical utility in the management of CNS involvement in acute leukemia.


Asunto(s)
Médula Ósea , Compuestos Bicíclicos Heterocíclicos con Puentes , Sulfonamidas , Humanos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacocinética , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Compuestos Bicíclicos Heterocíclicos con Puentes/líquido cefalorraquídeo , Compuestos Bicíclicos Heterocíclicos con Puentes/sangre , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacocinética , Sulfonamidas/líquido cefalorraquídeo , Sulfonamidas/sangre , Masculino , Persona de Mediana Edad , Femenino , Anciano , Médula Ósea/efectos de los fármacos , Adulto , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos/líquido cefalorraquídeo , Antineoplásicos/sangre , Espectrometría de Masas en Tándem , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/líquido cefalorraquídeo , Leucemia Mieloide Aguda/sangre , Anciano de 80 o más Años , Cromatografía Liquida , Leucemia/tratamiento farmacológico , Leucemia/líquido cefalorraquídeo , Leucemia/sangre , Adulto Joven
9.
Int J Nanomedicine ; 19: 8769-8778, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39220196

RESUMEN

Introduction: The tumor microenvironment (TME) of pancreatic cancer is highly immunosuppressive and characterized by a large number of cancer-associated fibroblasts, myeloid-derived suppressor cells, and regulatory T cells. Stimulator of interferon genes (STING) is an endoplasmic reticulum receptor that plays a critical role in immunity. STING agonists have demonstrated the ability to inflame the TME, reduce tumor burden, and confer anti-tumor activity in mouse models. 2'3' cyclic guanosine monophosphate adenosine monophosphate (2'3'-cGAMP) is a high-affinity endogenous ligand of STING. However, delivering cGAMP to antigen-presenting cells and tumor cells within the cytosol remains challenging due to membrane impermeability and poor stability. Methods: In this study, we encapsulated 2'3'-cGAMP in a lipid nanoparticle (cGAMP-LNP) designed for efficient cellular delivery. We assessed the properties of the nanoparticles using a series of in-vitro studies designed to evaluate their cellular uptake, cytosolic release, and minimal cytotoxicity. Furthermore, we examined the nanoparticle's anti-tumor effect in a syngeneic mouse model of pancreatic cancer. Results: The lipid platform significantly increased the cellular uptake of 2'3'-cGAMP. cGAMP-LNP exhibited promising antitumor activity in the syngeneic mouse model of pancreatic cancer. Discussion: The LNP platform shows promise for delivering exogenous 2'3'-cGAMP or its derivatives in cancer therapy.


Asunto(s)
Proteínas de la Membrana , Nanopartículas , Nucleótidos Cíclicos , Neoplasias Pancreáticas , Microambiente Tumoral , Animales , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Nanopartículas/química , Nanopartículas/administración & dosificación , Nucleótidos Cíclicos/farmacología , Nucleótidos Cíclicos/química , Nucleótidos Cíclicos/farmacocinética , Nucleótidos Cíclicos/administración & dosificación , Proteínas de la Membrana/agonistas , Ratones , Línea Celular Tumoral , Humanos , Microambiente Tumoral/efectos de los fármacos , Ratones Endogámicos C57BL , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Liposomas/química , Liposomas/farmacología , Liposomas/farmacocinética , Liposomas/administración & dosificación
10.
Clin Pharmacokinet ; 63(9): 1301-1312, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39243304

RESUMEN

BACKGROUND AND OBJECTIVE: Asciminib is approved in patients with Philadelphia chromosome-positive chronic myeloid leukemia in chronic phase (Ph+ CML-CP) treated with ≥ 2 prior tyrosine kinase inhibitors. Here, we aimed to demonstrate similarity in efficacy/safety of asciminib 80 mg once daily (q.d.) versus 40 mg twice daily (b.i.d.) in patients with CML-CP without T315I mutation and support the use of the 200-mg b.i.d. dosage in patients harboring T315I, using model-informed drug development. METHODS: Data were collected from 199 patients in the phase I (NCT02081378; 10-200 mg b.i.d. or 10-400 mg q.d.) and 154 patients in the phase III (NCT03106779; 40 mg b.i.d.) studies. Evaluations were based on population pharmacokinetics (PopPK) and exposure-response (efficacy/safety) analyses. RESULTS: PopPK showed comparable exposure (area under the curve, AUC0-24h) for 40 mg b.i.d. and 80 mg q.d. (12,638 vs 12,646 ng*h/mL); average maximum and minimum plasma concentrations for 80 mg q.d. were 1.61- and 0.72-fold those of 40 mg b.i.d., respectively. Exposure-response analyses predicted similar major molecular response rates for 40 mg b.i.d. and 80 mg q.d. (Week 24: 27.6% vs 24.8%; Week 48: 32.3% vs 30.6%). Results also established adequacy of 200 mg b.i.d. in patients with T315I mutation (Week 24: 20.7%; Week 48: 23.7%), along with a similar safety profile for all dose regimens. CONCLUSIONS: Similarity between 40 mg b.i.d. and 80 mg q.d. regimens was investigated, demonstrating similar and substantial efficacy with well-tolerated safety in patients without T315I mutation. The 200-mg b.i.d. dose was deemed safe and effective for patients with T315I mutation.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva , Mutación , Inhibidores de Proteínas Quinasas , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Masculino , Femenino , Persona de Mediana Edad , Adulto , Anciano , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/uso terapéutico , Cromosoma Filadelfia , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/genética , Relación Dosis-Respuesta a Droga , Antineoplásicos/farmacocinética , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Antineoplásicos/efectos adversos , Adulto Joven , Anciano de 80 o más Años , Área Bajo la Curva , Niacinamida/análogos & derivados , Pirazoles
11.
J Med Chem ; 67(18): 16056-16071, 2024 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-39230932

RESUMEN

The histone lysine methyltransferase NSD2 has been recognized as an attractive target for cancer treatment, due to the functional implication of its dysregulation in the initiation and progression of many cancers. Although considerable efforts have been made to develop NSD2 small-molecule inhibitors, highly potent and selective ones are still rarely available till now. Here, we report the discovery of a series of novel NSD2 inhibitors via an extensive SAR exploration of the privileged quinazoline scaffold within compound 8. The most promising compound 42 showed excellent NSD2 enzymatic inhibitory activity and good antiproliferative activity in cells. In addition, it demonstrated favorable pharmacokinetic properties and significantly inhibited the tumor growth in a RS411 tumor xenograft model with good safety. Taken together, compound 42 could be a promising NSD2 inhibitor and deserves further investigation.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Animales , Relación Estructura-Actividad , Quinazolinas/farmacología , Quinazolinas/química , Quinazolinas/síntesis química , Quinazolinas/farmacocinética , Ratones , Descubrimiento de Drogas , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Desnudos , Ratas
12.
J Med Chem ; 67(18): 16248-16269, 2024 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-39255403

RESUMEN

Given the considerable potential of DOT1LR231Q inhibitors in lung cancer therapy and the problematic pharmacokinetics of nucleoside inhibitors, our group launched a development program of non-nucleoside DOT1LR231Q inhibitors to improve the pharmacokinetic properties. Herein, two series of non-nucleoside compounds bearing piperidine or 3-(aminomethyl)pyrrolidin-3-ol as "ribose mimics" were designed and evaluated through antiproliferation assay and western blot analysis. The optimal TB22 inhibited the proliferation of H460R231Q cells with an IC50 value of 2.85 µM, about 13-fold more potent than SGC0946. Notably, TB22 demonstrated significant in vivo efficacy (TGI = 60.57%) in H460R231Q cell-derived xenograft models and improved pharmacokinetic properties (t1/2 = 6.06 ± 2.94 h and CL = 55.18 ± 8.56 mL/kg/min). Moreover, a mechanism study validated that TB22 suppressed malignant phenotypes of lung cancer cells harboring R231Q mutation via the MAPK/ERK signaling pathway. This work provides a promising molecule for lung cancer therapy in favor of clinical patients.


Asunto(s)
Antineoplásicos , Proliferación Celular , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Animales , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Antineoplásicos/química , Antineoplásicos/síntesis química , Proliferación Celular/efectos de los fármacos , Ratones , Línea Celular Tumoral , Relación Estructura-Actividad , Ratones Desnudos , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/uso terapéutico , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C , N-Metiltransferasa de Histona-Lisina
13.
Pharmacol Res ; 208: 107390, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39233056

RESUMEN

Glioma is one of the most common central nervous system (CNS) cancers that can be found within the brain and the spinal cord. One of the pressing issues plaguing the development of therapeutics for glioma originates from the selective and semipermeable CNS membranes: the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB). It is difficult to bypass these membranes and target the desired cancerous tissue because the purpose of the BBB and BSCB is to filter toxins and foreign material from invading CNS spaces. There are currently four varieties of Food and Drug Administration (FDA)-approved drug treatment for glioma; yet these therapies have limitations including, but not limited to, relatively low transmission through the BBB/BSCB, despite pharmacokinetic characteristics that allow them to cross the barriers. Steps must be taken to improve the development of novel and repurposed glioma treatments through the consideration of pharmacological profiles and innovative drug delivery techniques. This review addresses current FDA-approved glioma treatments' gaps, shortcomings, and challenges. We then outline how incorporating computational BBB/BSCB models and innovative drug delivery mechanisms will help motivate clinical advancements in glioma drug delivery. Ultimately, considering these attributes will improve the process of novel and repurposed drug development in glioma and the efficacy of glioma treatment.


Asunto(s)
Antineoplásicos , Barrera Hematoencefálica , Neoplasias Encefálicas , Sistemas de Liberación de Medicamentos , Desarrollo de Medicamentos , Glioma , Glioma/tratamiento farmacológico , Humanos , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Antineoplásicos/uso terapéutico , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología
14.
J Med Chem ; 67(19): 17000-17032, 2024 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-39283694

RESUMEN

TAM receptor tyrosine kinases have emerged as promising therapeutic targets for cancer treatment due to their roles in both tumor intrinsic survival mechanisms and suppression of antitumor immunity within the tumor microenvironment. Inhibiting MerTK and Axl selectively is believed to hinder cancer cell survival, reverse the protumor myeloid phenotype, and suppress efferocytosis, thereby eliciting an antitumor immune response. In this study, we present the discovery of A-910, a highly potent and selective dual MerTK/Axl inhibitor, achieved through a structure-based medicinal chemistry campaign. The lead compound exhibits favorable oral bioavailability, exceptional kinome selectivity, and significantly improved in vivo target engagement. These findings support the use of A-910 as an orally bioavailable in vivo tool compound for investigating the immunotherapy potential of dual MerTK/Axl inhibition.


Asunto(s)
Tirosina Quinasa del Receptor Axl , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras , Tirosina Quinasa c-Mer , Tirosina Quinasa c-Mer/antagonistas & inhibidores , Tirosina Quinasa c-Mer/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/administración & dosificación , Humanos , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Administración Oral , Relación Estructura-Actividad , Disponibilidad Biológica , Ratones , Descubrimiento de Drogas , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Ratas
15.
Int J Pharm ; 665: 124720, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39284421

RESUMEN

Triple-negative breast cancer (TNBC) has poor prognosis. Carboplatin (Crb) is a widely used chemotherapeutic agent, in TNBC but with serious systemic toxicity and poor tumor targeting. Bioinspired drug-loaded platelets (Plt) and Plt-coated nanocarriers evade macrophage phagocytosis by membrane proteins like CD47. The goal of this study was preparation of a novel alginate-poly (ß-amino ester) (PßAE) nanoparticles (NPs) for targeted delivery of Crb to TNBC cells by developing and comparison of two bioinspired carriers of Plt membrane (PltM) coated Crb-loaded alginate-poly (ß-amino ester) nanoparticles (PltM@Crb-PßAE-ALG NPs) and Plt loaded Crb (Plt@Crb). The NPs were prepared by ionic gelation and subsequently were coated by platelet membrane using ultra-sonication method. The loading efficiency, release profile, and in vitro cytotoxicity of both formulations were evaluated on HUVEC and 4 T1 cells. Additionally, the in vivo tumor targeting, therapeutic efficacy, and organ toxicity of the two formulations were assessed in a murine tumor model. Results showed both Plt@Crb and (PltM@Crb-PßAE-ALG NPs) exhibited high drug loading efficiency, sustained release, enhanced cytotoxicity against 4 T1 cells, and decreased cytotoxicity in normal cells (HUVEC) in vitro. In vivo studies revealed that although both formulations considerably improved tumor inhibition compared to free Crb, but the PltM@Crb-PßAE-ALG NPs demonstrated superior cytotoxicity and therapeutic efficacy, thanks to improved Crb's internalization efficiency, enhanced stability, and controlled release properties.


Asunto(s)
Alginatos , Antineoplásicos , Plaquetas , Carboplatino , Liberación de Fármacos , Células Endoteliales de la Vena Umbilical Humana , Nanopartículas , Polímeros , Animales , Alginatos/química , Alginatos/administración & dosificación , Femenino , Humanos , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Carboplatino/administración & dosificación , Carboplatino/química , Nanopartículas/química , Nanopartículas/administración & dosificación , Línea Celular Tumoral , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Polímeros/química , Portadores de Fármacos/química , Ratones , Polielectrolitos/química , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Ratones Endogámicos BALB C , Supervivencia Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología
16.
J Med Chem ; 67(19): 17520-17541, 2024 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-39340456

RESUMEN

Androgen receptor (AR) is an important therapeutic target for prostate cancer (PCa) treatment, but prolonged use of AR antagonists has led to variant drug-resistant mutations. Since all marketed AR antagonists target the ligand binding pocket (LBP) of AR, to mitigate cross-resistance, a new drug pocket named Dimer Interface Pocket was discovered and a novel AR antagonist M17-B15 was identified. M17-B15 showed strong in vitro efficacy against PCa but had poor pharmacokinetic properties in vivo. In this study, through rational design and structure-activity relationship exploration, a series of thiadiazoleamide derivatives represented by N29 (IC50 = 0.018 µM) were identified with dominant AR antagonistic activity and remarkable anti-PCa activity in vitro. Furthermore, N29 effectively inhibited a series of typical drug-resistant AR mutants. The improved oral bioavailability of N29 facilitated its efficacy via oral administration, significantly inhibiting LNCaP xenograft tumor in vivo, presenting a promising therapeutic application for PCa.


Asunto(s)
Antagonistas de Receptores Androgénicos , Neoplasias de la Próstata , Receptores Androgénicos , Tiadiazoles , Humanos , Masculino , Tiadiazoles/farmacología , Tiadiazoles/química , Tiadiazoles/farmacocinética , Tiadiazoles/síntesis química , Animales , Receptores Androgénicos/metabolismo , Antagonistas de Receptores Androgénicos/farmacología , Antagonistas de Receptores Androgénicos/química , Antagonistas de Receptores Androgénicos/farmacocinética , Antagonistas de Receptores Androgénicos/síntesis química , Administración Oral , Relación Estructura-Actividad , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Línea Celular Tumoral , Ratones , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Ratones Desnudos , Amidas/química , Amidas/farmacología , Amidas/síntesis química , Amidas/farmacocinética , Descubrimiento de Drogas , Ensayos Antitumor por Modelo de Xenoinjerto , Disponibilidad Biológica , Ratas
17.
J Med Chem ; 67(19): 17053-17069, 2024 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-39348183

RESUMEN

c-Met is an attractive therapeutic target in multiple tumors. Previous studies have discovered some effective proteolysis-targeting chimeras (PROTACs) able to degrade c-Met; however, the structure-activity relationship (SAR), degradation selectivity, and pharmacokinetic profiles of c-Met PROTACs have, to date, remained largely unknown. Herein, through extensive SAR studies on various warheads, linkers, and E3 ligase ligands, a novel potent c-Met PROTAC Met-DD4 was identified. Our results suggested that Met-DD4 could induce robust c-Met degradation with excellent selectivity (DC50 = 6.21 nM), substantially killing the c-Met-addicted cancer cells (IC50 = 4.37 nM). Furthermore, in vivo studies showed that Met-DD4 could achieve excellent oral bioavailability and c-Met degradation, strongly retarding tumor growth with minute organ toxicity. Overall, this study reveals that targeted degradation of c-Met is a promising strategy for the treatment of c-Met-addicted cancers and provides novel lead compounds for the clinical translation of c-Met PROTACs.


Asunto(s)
Antineoplásicos , Disponibilidad Biológica , Proteolisis , Proteínas Proto-Oncogénicas c-met , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Humanos , Proteolisis/efectos de los fármacos , Relación Estructura-Actividad , Animales , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Antineoplásicos/síntesis química , Administración Oral , Línea Celular Tumoral , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Ratones Desnudos , Proliferación Celular/efectos de los fármacos , Quimera Dirigida a la Proteólisis
18.
Arch Microbiol ; 206(10): 403, 2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39276253

RESUMEN

Seaweed endophytes are a rich source of microbial diversity and bioactive compounds. This review provides a comprehensive analysis of the microbial diversity associated with seaweeds and their interaction between them. These diverse bacteria and fungi have distinct metabolic pathways, which result in the synthesis of bioactive compounds with potential applications in a variety of health fields. We examine many types of seaweed-associated microorganisms, their bioactive metabolites, and their potential role in cancer treatment using a comprehensive literature review. By incorporating recent findings, we hope to highlight the importance of seaweed endophytes as a prospective source of novel anticancer drugs and promote additional studies in this area. We also investigate the pharmacokinetic and pharmacodynamic profiles of these bioactive compounds because understanding their absorption, distribution, metabolism, excretion (ADMET), and toxicity profiles is critical for developing bioactive compounds with anticancer potential into effective cancer drugs. This knowledge ensures the safety and efficacy of proposed medications prior to clinical trials. This study not only provides promise for novel and more effective treatments for cancer with fewer side effects, but it also emphasizes the necessity of sustainable harvesting procedures and ethical considerations for protecting the delicate marine ecology during bioprospecting activities.


Asunto(s)
Antineoplásicos , Bacterias , Endófitos , Hongos , Algas Marinas , Algas Marinas/química , Endófitos/metabolismo , Endófitos/química , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Humanos , Hongos/efectos de los fármacos , Hongos/metabolismo , Bacterias/efectos de los fármacos , Bacterias/metabolismo , Neoplasias/tratamiento farmacológico , Animales , Biodiversidad
19.
Ther Deliv ; 15(10): 749-768, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39287183

RESUMEN

Aim: Dasatinib (DST) is an oral tyrosine kinase inhibitor with poor aqueous solubility. To outwit this issue, a solid self-nano emulsifying drug delivery system (S-SNEDDS) of DST was formulated.Methods: I-optimal mixture design was used for optimization of DST-loaded SNEDDS using Linalool, Cremophor RH40 and Transcutol P. S-SNEDDS underwent physicochemical characterization, in-vitro release and ex-vivo permeation, cell-based assays and pharmacokinetic study.Results: DST-S-SNEDDS showed globule size and PDI of 141.53 ± 5.371 nm and 0.282 ± 0.020, respectively. DST-S-SNEDDS revealed significantly lower IC50 (1.825 µg/mL) than free DST (7.298 µg/mL) in MDA-MB-231. In-vivo pharmacokinetic study revealed 1.94-fold increment in AUC0-t for the DST-S-SNEDDS group than free DST.Conclusion: S-SNEDDS could be promising approach for improving bioavailability and efficacy of DST.


[Box: see text].


Asunto(s)
Disponibilidad Biológica , Dasatinib , Emulsiones , Solubilidad , Dasatinib/farmacocinética , Dasatinib/administración & dosificación , Dasatinib/química , Animales , Humanos , Línea Celular Tumoral , Tamaño de la Partícula , Liberación de Fármacos , Polietilenglicoles/química , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Sistemas de Liberación de Medicamentos/métodos , Ratas Sprague-Dawley , Nanopartículas/química , Ratas , Sistema de Administración de Fármacos con Nanopartículas/química , Masculino , Glicoles de Etileno/química , Administración Oral , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos/química , Química Farmacéutica/métodos
20.
AAPS PharmSciTech ; 25(7): 221, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39317842

RESUMEN

This study aims to enhance the solubility of Olaparib, classified as biopharmaceutical classification system (BCS) class IV due to its low solubility and bioavailability using a solid self-nanoemulsifying drug delivery system (S-SNEDDS). For this purpose, SNEDDS formulations were created using Capmul MCM as the oil, Tween 80 as the surfactant, and PEG 400 as the co-surfactant. The SNEDDS formulation containing olaparib (OLS-352), selected as the optimal formulation, showed a mean droplet size of 87.0 ± 0.4 nm and drug content of 5.53 ± 0.09%. OLS-352 also demonstrated anticancer activity against commonly studied ovarian (SK-OV-3) and breast (MCF-7) cancer cell lines. Aerosil® 200 and polyvinylpyrrolidone (PVP) K30 were selected as solid carriers, and S-SNEDDS formulations were prepared using the spray drying method. The drug concentration in S-SNEDDS showed no significant changes (98.4 ± 0.30%, 25℃) with temperature fluctuations during the 4-week period, demonstrating improved storage stability compared to liquid SNEDDS (L-SNEDDS). Dissolution tests under simulated gastric and intestinal conditions revealed enhanced drug release profiles compared to those of the raw drug. Additionally, the S-SNEDDS formulation showed a fourfold greater absorption in the Caco-2 assay than the raw drug, suggesting that S-SNEDDS could improve the oral bioavailability of poorly soluble drugs like olaparib, thus enhancing therapeutic outcomes. Furthermore, this study holds significance in crafting a potent and cost-effective pharmaceutical formulation tailored for the oral delivery of poorly soluble drugs.


Asunto(s)
Disponibilidad Biológica , Sistemas de Liberación de Medicamentos , Emulsiones , Ftalazinas , Piperazinas , Solubilidad , Piperazinas/química , Piperazinas/administración & dosificación , Piperazinas/farmacocinética , Humanos , Ftalazinas/química , Ftalazinas/administración & dosificación , Ftalazinas/farmacocinética , Ftalazinas/farmacología , Emulsiones/química , Sistemas de Liberación de Medicamentos/métodos , Línea Celular Tumoral , Estabilidad de Medicamentos , Química Farmacéutica/métodos , Tamaño de la Partícula , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos/química , Antineoplásicos/farmacología , Tensoactivos/química , Portadores de Fármacos/química , Polietilenglicoles/química , Células MCF-7 , Liberación de Fármacos , Nanopartículas/química , Composición de Medicamentos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA