Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Small ; 19(2): e2202343, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36394151

RESUMEN

Ectopic pregnancy (EP) is the leading cause of maternity-related death in the first trimester of pregnancy. Approximately 98% of ectopic implantations occur in the fallopian tube, and expedient management is crucial for preventing hemorrhage and maternal death in the event of tubal rupture. Current ultrasound strategies misdiagnose EP in up to 40% of cases, and the failure rate of methotrexate treatment for confirmed EP exceeds 10%. Here the first theranostic strategy for potential management of EP is reported using a near-infrared naphthalocyanine dye encapsulated within polymeric nanoparticles. These nanoparticles preferentially accumulate in the developing murine placenta within 24 h following systemic administration, and enable visualization of implantation sites at various gestational stages via fluorescence and photoacoustic imaging. These nanoparticles do not traverse the placental barrier to the fetus or impact fetal development. However, excitation of nanoparticles localized in specific placentas with focused NIR light generates heat (>43 °C) sufficient for disruption of placental function, resulting in the demise of targeted fetuses with no effect on adjacent fetuses. This novel approach would enable diagnostic confirmation of EP when current imaging strategies are unsuccessful, and elimination of EP could subsequently be achieved using the same nano-agent to generate localized hyperthermia resulting in targeted placental impairment.


Asunto(s)
Hipertermia Inducida , Embarazo Ectópico , Embarazo , Femenino , Humanos , Animales , Ratones , Placenta/diagnóstico por imagen , Embarazo Ectópico/terapia , Trompas Uterinas/diagnóstico por imagen , Ultrasonografía
2.
Small ; : e2300011, 2023 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-37452434

RESUMEN

Patients undergoing gynecological procedures suffer from lasting side effects due to intraoperative nerve damage. Small, delicate nerves with complex and nonuniform branching patterns in the female pelvic neuroanatomy make nerve-sparing efforts during standard gynecological procedures such as hysterectomy, cystectomy, and colorectal cancer resection difficult, and thus many patients are left with incontinence and sexual dysfunction. Herein, a near-infrared (NIR) fluorescent nerve-specific contrast agent, LGW08-35, that is spectrally compatible with clinical fluorescence guided surgery (FGS) systems is formulated and characterized for rapid implementation for nerve-sparing gynecologic surgeries. The toxicology, pharmacokinetics (PK), and pharmacodynamics (PD) of micelle formulated LGW08-35 are examined, enabling the determination of the optimal imaging doses and time points, blood and tissue uptake parameters, and maximum tolerated dose (MTD). Application of the formulated fluorophore to imaging of female rat and swine pelvic neuroanatomy validates the continued clinical translation and use for real-time identification of important nerves such as the femoral, sciatic, lumbar, iliac, and hypogastric nerves. Further development of LGW08-35 for clinical use will unlock a valuable tool for surgeons in direct visualization of important nerves and contribute to the ongoing characterization of the female pelvic neuroanatomy to eliminate the debilitating side effects of nerve damage during gynecological procedures.

3.
Mol Pharm ; 19(8): 2690-2711, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33605146

RESUMEN

The field of nanomedicine continues to grow with new technologies and formulations in development for several disease states. Much research focuses on the use of injectable nanomedicines for treatment of neoplasms; however, there are several formulations in development that use nanotechnology that can be administered enterally for noncancer indications. These nanomedicine treatments have been developed for systemic drug delivery or local drug delivery along the gastrointestinal tract. This Review gives a brief overview of the alimentary canal and highlights new research in nanomedicine in noncancer disease states delivered via enteral routes of administration. Relevant recent research is summarized on the basis of the targeted site of action or absorption, including the buccal, sublingual, stomach, small intestine, and large intestine areas of the alimentary canal. The benefits of nanodrug delivery are discussed as well as barriers and challenges for future development in the field.


Asunto(s)
Nanomedicina , Neoplasias , Sistemas de Liberación de Medicamentos , Tracto Gastrointestinal , Humanos , Nanotecnología
4.
Mol Pharm ; 19(12): 4696-4704, 2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36409995

RESUMEN

Recently, therapeutics based on mRNA (mRNA) have attracted significant interest for vaccines, cancer immunotherapy, and gene editing. However, the lack of biocompatible vehicles capable of delivering mRNA to the target tissue and efficiently expressing the encoded proteins impedes the development of mRNA-based therapies for a variety of diseases. Herein, we report mRNA-loaded polymeric nanoparticles based on diethylenetriamine-substituted poly(aspartic acid) that induce protein expression in the lungs and muscles following intravenous and intramuscular injections, respectively. Animal studies revealed that the amount of polyethylene glycol (PEG) on the nanoparticle surface affects the translation of the delivered mRNA into the encoded protein in the target tissue. After systemic administration, only mRNA-loaded nanoparticles modified with PEG at a molar ratio of 1:1 (PEG/polymer) induce protein expression in the lungs. In contrast, protein expression was detected only following intramuscular injection of mRNA-loaded nanoparticles with a PEG/polymer ratio of 10:1. These findings suggest that the PEG density on the surface of poly(aspartic acid)-based nanoparticles should be optimized for different delivery routes depending on the purpose of the mRNA treatment.


Asunto(s)
Ácido Aspártico , Nanopartículas , Animales , ARN Mensajero/genética , Polímeros , Inmunoterapia , Polietilenglicoles
5.
Small ; 16(18): e1906936, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32250034

RESUMEN

Endometriosis is a painful disorder where endometrium-like tissue forms lesions outside of the uterine cavity. Intraoperative identification and removal of these lesions are difficult. This study presents a nanoplatform that concurrently delineates and ablates endometriosis tissues using real-time near-infrared (NIR) fluorescence and photothermal therapy (PTT). The nanoplatform consists of a dye, silicon naphthalocyanine (SiNc), capable of both NIR fluorescence imaging and PTT, and a polymeric nanoparticle as a SiNc carrier to endometriosis tissue following systemic administration. To achieve high contrast during fluorescence imaging of endometriotic lesions, nanoparticles are constructed to be non-fluorescent prior to internalization by endometriosis cells. In vitro studies confirm that these nanoparticles activate the fluorescence signal following internalization in macaque endometrial stromal cells and ablate them by increasing cellular temperature to 53 ° C upon interaction with NIR light. To demonstrate in vivo efficiency of the nanoparticles, biopsies of endometrium and endometriosis from rhesus macaques are transplanted into immunodeficient mice. Imaging with the intraoperative Fluobeam 800 system reveals that 24 h following intravenous injection, nanoparticles efficiently accumulate in, and demarcate, endometriotic grafts with fluorescence. Finally, the nanoparticles increase the temperature of endometriotic grafts up to 47 °C upon exposure to NIR light, completely eradicating them after a single treatment.


Asunto(s)
Endometriosis , Hipertermia Inducida , Nanopartículas , Fototerapia , Animales , Endometriosis/diagnóstico por imagen , Endometriosis/terapia , Femenino , Humanos , Macaca mulatta , Ratones , Imagen Óptica
6.
Nanomedicine ; 19: 39-48, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31022465

RESUMEN

This work looks to improve the efficacy of Adriamycin (ADR) while mitigating its cardiotoxicity using combinations of micellar resveratrol (R): quercetin (Q) (mRQ) or R: curcumin (C) (mRC) in healthy mice and ovarian cancer xenograft models. Ovarian cancer cells, ES2-Luc, or A2780ADR are inoculated in mice (n =4/group) and sorted into eight cohorts. Mice are treated weekly for 4 weeks with ADR, ADR+mRQ, ADR+mRC, or controls (saline, empty micelles, ADR+EM, mRQ, or mRC). To evaluate the degree of cardioprotection, serum is collected to determine the cardiac Troponin I (cTnI). Cardiac tissue is collected for morphological evaluation and evaluation of creatine kinase levels. Our results indicate that mRQ+ADR is statistically significant in tumor reduction in xenograft models. In healthy mice, the left ventricular ejection fraction and fractional shortening in the ADR treated group is most compromised. Co-administration of mRQ with ADR can reduce ADR dosing through chemosensitization while being cardioprotective.


Asunto(s)
Cardiotoxicidad/tratamiento farmacológico , Curcumina/uso terapéutico , Doxorrubicina/efectos adversos , Micelas , Neoplasias Ováricas/tratamiento farmacológico , Polímeros/química , Quercetina/uso terapéutico , Resveratrol/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Cardiotoxicidad/diagnóstico por imagen , Cardiotoxicidad/fisiopatología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Curcumina/administración & dosificación , Curcumina/farmacología , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Concentración 50 Inhibidora , Mediciones Luminiscentes , Ratones , Neoplasias Ováricas/diagnóstico por imagen , Quercetina/administración & dosificación , Quercetina/farmacología , Resveratrol/administración & dosificación , Resveratrol/farmacología , Volumen Sistólico/efectos de los fármacos , Troponina I/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Invest New Drugs ; 34(1): 24-40, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26563191

RESUMEN

Coibamide A is a cytotoxic lariat depsipeptide isolated from a rare cyanobacterium found within the marine reserve of Coiba National Park, Panama. Earlier testing of coibamide A in the National Cancer Institute in vitro 60 human tumor cell line panel (NCI-60) revealed potent anti-proliferative activity and a unique selectivity profile, potentially reflecting a new target or mechanism of action. In the present study we evaluated the antitumor activity of coibamide A in several functional cell-based assays and in vivo. U87-MG and SF-295 glioblastoma cells showed reduced migratory and invasive capacity and underwent G1 cell cycle arrest as, likely indirect, consequences of treatment. Coibamide A inhibited extracellular VEGFA secreted from U87-MG glioblastoma and MDA-MB-231 breast cancer cells with low nM potency, attenuated proliferation and migration of normal human umbilical vein endothelial cells (HUVECs) and selectively decreased expression of vascular endothelial growth factor receptor 2 (VEGFR2). We report that coibamide A retains potent antitumor properties in a nude mouse xenograft model of glioblastoma; established subcutaneous U87-MG tumors failed to grow for up to 28 days in response to 0.3 mg/Kg doses of coibamide A. However, the natural product was also associated with varied patterns of weight loss and thus targeted delivery and/or medicinal chemistry approaches will almost certainly be required to improve the toxicity profile of this unusual macrocycle. Finally, similarities between coibamide A- and apratoxin A-induced changes in cell morphology, decreases in VEGFR2 expression and macroautophagy signaling in HUVECs raise the possibility that both cyanobacterial natural products share a common mechanism of action.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Depsipéptidos/farmacología , Glioblastoma/tratamiento farmacológico , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Glioblastoma/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Desnudos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Pharm ; 13(6): 2070-83, 2016 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-27170529

RESUMEN

We report an efficient therapeutic modality for platinum resistant ovarian cancer based on siRNA-mediated suppression of a multifunctional DJ-1 protein that is responsible for the proliferation, growth, invasion, oxidative stress, and overall survival of various cancers. The developed therapeutic strategy can work alone or in concert with a low dose of the first line chemotherapeutic agent cisplatin, to elicit a maximal therapeutic response. To achieve an efficient DJ-1 knockdown, we constructed the polypropylenimine dendrimer-based nanoplatform targeted to LHRH receptors overexpressed on ovarian cancer cells. The quantitative PCR and Western immunoblotting analysis revealed that the delivered DJ-1 siRNA downregulated the expression of targeted mRNA and corresponding protein by more than 80% in various ovarian cancer cells. It was further demonstrated that siRNA-mediated DJ-1 suppression dramatically impaired proliferation, viability, and migration of the employed ovarian cancer cells. Finally, the combinatorial approach led to the most pronounced therapeutic response in all the studied cell lines, outperforming both siRNA-mediated DJ-1 knockdown and cisplatin treatment alone. It is noteworthy that the platinum-resistant cancer cells (A2780/CDDP) with the highest basal level of DJ-1 protein are most susceptible to the developed therapy and this susceptibility declines with decreasing basal levels of DJ-1. Finally, we interrogate the molecular underpinnings of the DJ-1 knockdown effects in the treatment of the ovarian cancer cells. By using various experimental techniques, it was revealed that DJ-1 depletion (1) decreases the activity of the Akt pathway, thereby reducing cellular proliferation and migration and increasing the antiproliferative effect of cisplatin on ovarian cancer cells; (2) enhances the activity of p53 tumor suppressor protein therefore restoring cell cycle arrest functionality and upregulating the Bax-caspase pathway, triggering cell death; and (3) weakens the cellular defense mechanisms against inherited oxidative stress thereby increasing toxic intracellular radicals and amplifying the reactive oxygen species created by the administration of cisplatin.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Compuestos Organoplatinos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Proteína Desglicasa DJ-1/metabolismo , ARN Interferente Pequeño/administración & dosificación , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dendrímeros/administración & dosificación , Femenino , Humanos , Nanomedicina/métodos , Neoplasias Ováricas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores LHRH/metabolismo
9.
Mol Pharm ; 12(12): 4386-94, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26485440

RESUMEN

Nerve damage during surgery is a common morbidity experienced by patients that leaves them with chronic pain and/or loss of function. Currently, no clinically approved imaging technique exists to enhance nerve visualization in the operating room. Fluorescence image-guided surgery has gained in popularity and clinical acceptance over the past decade with a handful of imaging systems approved for clinical use. However, contrast agent development to complement these fluorescence-imaging systems has lagged behind with all currently approved fluorescent agents providing untargeted blood pool information. Nerve-specific fluorophores are known, however translations of these agents to the clinic has been complicated by their lipophilic nature, which necessitates specialized formulation strategies for successful systemic administration. To date the known nerve-specific fluorophores have only been demonstrated preclinically due to the necessity of a dimethyl sulfoxide containing formulation for solubilization. In the current study, a polymeric micellar (PM) formulation strategy was developed for a representative nerve-specific fluorophore from the distyrylbenzene family, BMB. The PM formulation strategy was able to solubilize BMB and demonstrated improved nerve-specific accumulation and fluorescence intensity when the same fluorophore dose was administered to mice utilizing the previous formulation strategy. The success of the PM formulation strategy will be important for moving toward clinical translation of these novel nerve-specific probes as it is nontoxic and biodegradable and has the potential to decrease the necessary dose for imaging while also improving the safety profile.


Asunto(s)
Portadores de Fármacos/química , Colorantes Fluorescentes/química , Polímeros/química , Animales , Línea Celular , Línea Celular Tumoral , Química Farmacéutica/métodos , Medios de Contraste/química , Dimetilsulfóxido/química , Sistemas de Liberación de Medicamentos/métodos , Humanos , Masculino , Ratones , Micelas , Estirenos/química
10.
Pharm Res ; 31(3): 660-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24065590

RESUMEN

PURPOSE: The in vitro inhibitory effect of Docetaxel (DTX) and Everolimus (EVR) alone and together in poly(ethylene glycol)-block-poly(D,L-lactic acid) (PEG-b-PLA) nanocarriers on angiogenic processes and acute toxicity in mice was evaluated. METHODS: PEG-b-PLA DTX and/or EVR nanocarriers were characterized for size, drug loading, stability, and drug release. Cell proliferation, tubule formation, and migration studies were performed in Human Umbilical Vein Endothelial Cells (HUVEC) and Maximum Tolerated Doses (MTD) studies were in mice. RESULTS: DTX and EVR loading was 1.93 and 2.00 mg/mL respectively with similar solubilities for dual-drug micelles. All micelles were below 30 nm with diffusion controlled drug release. The IC50 for DTX, EVR micelles were, 6.80 ± 0.67, 18.57 ± 2.86 and 0.65 ± 0.11 nM respectively with a synergistic inhibitory effect for dual-drug nanocarriers. Significant inhibition of tube formation occurred upon treatment with dual-drug nanocarriers as compared to individual micelles. EVR presence in dual-drug nanocarriers was able to significantly increase the inhibition of the migration of HUVEC by DTX. The MTDs for EVR, DTX and dual-drug micelles were 50, 30 and 20 mg/kg for each respectively. CONCLUSIONS: DTX-EVR dual-drug nanocarriers have antiangiogenic effects in vitro mediated through cellular angiogenic process and possess clinically relevant MTD.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Portadores de Fármacos/química , Lactatos/química , Polietilenglicoles/química , Sirolimus/análogos & derivados , Taxoides/administración & dosificación , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/toxicidad , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Docetaxel , Portadores de Fármacos/toxicidad , Everolimus , Femenino , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Lactatos/toxicidad , Ratones , Micelas , Polietilenglicoles/toxicidad , Sirolimus/administración & dosificación , Sirolimus/farmacología , Sirolimus/toxicidad , Taxoides/farmacología , Taxoides/toxicidad , Pruebas de Toxicidad Aguda
11.
Adv Mater ; 36(16): e2304724, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37653576

RESUMEN

Fluorescence-guided surgery (FGS) is poised to revolutionize surgical medicine through near-infrared (NIR) fluorophores for tissue- and disease-specific contrast. Clinical open and laparoscopic FGS vision systems operate nearly exclusively at NIR wavelengths. However, tissue-specific NIR contrast agents compatible with clinically available imaging systems are lacking, leaving nerve tissue identification during prostatectomy a persistent challenge. Here, it is shown that combining drug-like molecular design concepts and fluorophore chemistry enabled the production of a library of NIR phenoxazine-based fluorophores for intraoperative nerve-specific imaging. The lead candidate readily delineated prostatic nerves in the canine and iliac plexus in the swine using the clinical da Vinci Surgical System that has been popularized for minimally invasive prostatectomy procedures. These results demonstrate the feasibility of molecular engineering of NIR nerve-binding fluorophores for ready integration into the existing surgical workflow, paving the path for clinical translation to reduce morbidity from nerve injury for prostate cancer patients.


Asunto(s)
Tejido Nervioso , Oxazinas , Neoplasias de la Próstata , Masculino , Humanos , Animales , Perros , Porcinos , Colorantes Fluorescentes/química , Prostatectomía/métodos
12.
Mol Pharm ; 10(5): 2071-8, 2013 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-23590802

RESUMEN

Angiogenesis is an essential process for disease progression in many solid tumors. There are several major cascade events in the angiogenic process that can be targeted to inhibit new blood vessel formation in the tumor tissue. The purpose of this work is to evaluate the inhibitory effect of paclitaxel (PTX) and rapamycin (RAP) as individual and in dual drug-loaded poly(ethylene glycol)-block-poly(d,l-lactic acid) (PEG-b-PLA) micelles on the angiogenic cascade processes of proliferation, migration, and tube formation. PEG-b-PLA PTX and/or RAP micelles were formed and characterized for size and drug loading. Sizes of individual and dual drug micelles were below 40 nm. PEG-b-PLA micelles significantly enhanced the aqueous solubility of PTX 1.80 mg/mL and RAP 1.60 mg/mL. The PTX-RAP dual drug PEG-b-PLA micelles were able to load PTX and RAP at 1.60 mg/mL for both drugs. Cell proliferation, apoptosis, tubule formation, and migration studies were performed in human umbilical vein endothelial cells (HUVEC). PTX and RAP in DMSO inhibited HUVEC proliferation with IC50 values of 0.82 ± 0.02 and 13 829 ± 681 nM, respectively, while the combination of both drugs in DMSO produced synergistic inhibition. PTX and RAP individual micelles had IC50 values of 6.3 ± 1.1 and 14 051 ± 821 nM, respectively. PTX and dual drug micelles had a synergistic inhibition effect on HUVEC proliferation through the induction of apoptosis via caspase 3/7 activity. In vitro tube formation assay demonstrated significant inhibition of tube formation upon treatment with dual drug micelles as compared to individual PTX or RAP micelles. Migration studies in HUVEC have shown that individual PTX micelles inhibited cell migration at 1 nM, while RAP micelles did not show any inhibitory effect on cell migration. Interestingly, the presence of RAP in the dual drug micelles was able to initiate the inhibition of the migration of HUVEC at 0.1 nM concentration of PTX. These results indicate that PTX-RAP dual drug micelles have antiangiogenic effects in vitro mediated through three major events in the angiogenic process and have strong potential for further development as antiangiogenic chemotherapy.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Neovascularización Patológica/prevención & control , Paclitaxel/administración & dosificación , Sirolimus/administración & dosificación , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Sinergismo Farmacológico , Células Endoteliales de la Vena Umbilical Humana , Humanos , Micelas , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Tamaño de la Partícula , Polietilenglicoles/química
13.
Biomaterials ; 284: 121490, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35395454

RESUMEN

Iatrogenic nerve injury significantly affects surgical outcomes. Although intraoperative neuromonitoring is utilized, nerve identification remains challenging and the success of nerve sparing is strongly correlated with surgeon experience levels. Fluorescence guided surgery (FGS) offers a potential solution for improved nerve sparing by providing direct visualization of nerve tissue intraoperatively. However, novel probes for FGS face a long regulatory pathway to achieve clinical translation. Herein, we report on the development of a clinically-viable, gel-based formulation that enables direct administration of nerve-specific probes for nerve sparing FGS applications, facilitating clinical translation via the exploratory investigational new drug (eIND) guidance. The developed formulation possesses unique gelling characteristics, allowing it to be easily spread as a liquid followed by rapid gelling for subsequent tissue hold. Optimization of the direct administration protocol with our gel-based formulation enabled a total staining time of 1-2 min for compatibility with surgical procedures and successful clinical translation.


Asunto(s)
Colorantes Fluorescentes , Tejido Nervioso , Geles , Humanos , Enfermedad Iatrogénica
14.
Mol Pharm ; 8(4): 1257-65, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21630670

RESUMEN

Poly(ethylene glycol)-block-poly(D,L-lactic acid) (PEG-b-PLA) micelles have a proven capacity for drug solubilization and have entered phase III clinical trials as a substitute for Cremophor EL in the delivery of paclitaxel in cancer therapy. PEG-b-PLA is less toxic than Cremophor EL, enabling a doubling of paclitaxel dose in clinical trials. We show that PEG-b-PLA micelles act as a 3-in-1 nanocontainer for paclitaxel, 17-allylamino-17-demethoxygeldanamycin (17-AAG), and rapamycin for multiple drug solubilization. 3-in-1 PEG-b-PLA micelles were ca. 40 nm in diameter; dissolved paclitaxel, 17-AAG, and rapamycin in water at 9.0 mg/mL; and were stable for 24 h at 25 °C. The half-life for in vitro drug release (t(1/2)) for 3-in-1 PEG-b-PLA micelles was 1-15 h under sink conditions and increased in the order of 17-AAG, paclitaxel, and rapamycin. The t(1/2) values correlated with log P(o/w) values, implicating a diffusion-controlled mechanism for drug release. The IC(50) value of 3-in-1 PEG-b-PLA micelles for MCF-7 and 4T1 breast cancer cell lines was 114 ± 10 and 25 ± 1 nM, respectively; combination index (CI) analysis showed that 3-in-1 PEG-b-PLA micelles exert strong synergy in MCF-7 and 4T1 breast cancer cell lines. Notably, concurrent intravenous (iv) injection of paclitaxel, 17-AAG, and rapamycin using 3-in-1 PEG-b-PLA micelles was well-tolerated by FVB albino mice. Collectively, these results suggest that PEG-b-PLA micelles carrying paclitaxel, 17-AAG, and rapamycin will provide a simple yet safe and efficacious 3-in-1 nanomedicine for cancer therapy.


Asunto(s)
Micelas , Polímeros/química , Agua/química , Benzoquinonas/química , Línea Celular Tumoral , Proteínas HSP90 de Choque Térmico/química , Humanos , Lactamas Macrocíclicas/química , Ácido Láctico/química , Modelos Teóricos , Paclitaxel/química , Poliésteres , Polietilenglicoles/química , Sirolimus/química , Solubilidad , Serina-Treonina Quinasas TOR/química
15.
Artículo en Inglés | MEDLINE | ID: mdl-36053248

RESUMEN

Nerve damage is a major complication of surgery, causing pain and loss of function. We have identified novel near-infrared nerve-specific fluorophores that provide excellent nerve contrast with the ability to identify buried nerve tissue.

16.
Adv Drug Deliv Rev ; 177: 113957, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34481032

RESUMEN

Non-oral long-acting drug delivery systems (LADDS) encompass a range of technologies for precisely delivering drug molecules into target tissues either through the systemic circulation or via localized injections for treating chronic diseases like diabetes, cancer, and brain disorders as well as for age-related eye diseases. LADDS have been shown to prolong drug release from 24 h up to 3 years depending on characteristics of the drug and delivery system. LADDS can offer potentially safer, more effective, and patient friendly treatment options compared to more invasive modes of drug administration such as repeated injections or minor surgical intervention. Whilst there is no single technology or definition that can comprehensively embrace LADDS; for the purposes of this review, these systems include solid implants, inserts, transdermal patches, wafers and in situ forming delivery systems. This review covers common chronic illnesses, where candidate drugs have been incorporated into LADDS, examples of marketed long-acting pharmaceuticals, as well as newly emerging technologies, used in the fabrication of LADDS.


Asunto(s)
Enfermedad Crónica/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Polímeros/administración & dosificación , Animales , Formas de Dosificación , Humanos
17.
Adv Ther (Weinh) ; 4(7)2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34423111

RESUMEN

Nerves are extremely difficult to identify and are often accidently damaged during surgery, leaving patients with lasting pain and numbness. Herein, a novel near-infrared (NIR) nerve-specific fluorophore, LGW01-08, was utilized for enhanced nerve identification using fluorescence guided surgery (FGS), formulated using clinical translatable strategies. Formulated LGW01-08 was examined for toxicology, pharmacokinetics (PK), and pharmacodynamics (PD) parameters in preparation for future clinical translation. Optimal LGW01-08 imaging doses were identified in each formulation resulting in a 10x difference between the toxicity to imaging dose window. Laparoscopic swine surgery completed using the da Vinci surgical robot (Intuitive Surgical) demonstrated the efficacy of formulated LGW01-08 for enhanced nerve identification. NIR fluorescence imaging enabled clear identification of nerves buried beneath ~3 mm of tissue that were unidentifiable by white light imaging. These studies provide a strong basis for future clinical translation of NIR nerve-specific fluorophores for utility during FGS to improve patient outcomes.

18.
Pharm Res ; 27(11): 2421-32, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20700632

RESUMEN

PURPOSE: To prepare mixed polymeric micelles that can carry two different drugs, doxorubicin (DOX) and 17-hydroxyethylamino-17-demethoxygeldanamycin (GDM-OH), for combination cancer chemotherapy. METHODS: The pH-sensitive micelles were prepared from poly(ethylene glycol)-poly(aspartate hydrazide) block copolymers to which either DOX or GDM-OH is conjugated through acid-labile hydrazone bond (individual micelles). Mixed micelles were formed not only by simply mixing two different individual micelles in aqueous solutions (aqueous mixed micelles) but also by evaporating organic solvents from the organic/aqueous mixed solvents in which two block copolymers possessing different drugs were dissolved homogeneously (organic mixed micelles). Particle size measurements, pH-dependent drug release tests, cytotoxicity assays and western blot analysis were subsequently conducted. RESULTS: Individual and aqueous/organic mixed micelles showed clinically relevant particle size (<100 nm) and pH-dependent drug release patterns. Mixed polymer micelles suppress cancer cell growth effectively in a drug concentration, mixing method and schedule-dependent way. CONCLUSION: Combination chemotherapy using polymeric micelles seems to minimize a schedule-dependent change in combination drug efficacy in comparison to drug combination using DMSO formulations.


Asunto(s)
Portadores de Fármacos , Concentración de Iones de Hidrógeno , Micelas , Polímeros , Western Blotting , Línea Celular Tumoral , Humanos , Espectroscopía de Resonancia Magnética
19.
Adv Drug Deliv Rev ; 144: 16-34, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31461662

RESUMEN

Although many solid tumors use the lymphatic system to metastasize, there are few treatment options that directly target cancer present in the lymphatic system, and those that do are highly invasive, uncomfortable, and/or have limitations. In this review we provide a brief overview of lymphatic function and anatomy, discusses changes that befall the lymphatics in cancer and the mechanisms by which these changes occur, and highlight limitations of lymphatic drug delivery. We then go on to summarize relevant techniques and new research for targeting cancer populations in the lymphatics and enhancing drug delivery intralymphatically, including intralymphatic injections, isolated limb perfusion, passive nano drug delivery systems, and actively targeted nanomedicine.


Asunto(s)
Sistemas de Liberación de Medicamentos , Metástasis Linfática/tratamiento farmacológico , Vasos Linfáticos/metabolismo , Neoplasias/tratamiento farmacológico , Animales , Humanos , Neoplasias/metabolismo
20.
J Control Release ; 307: 272-281, 2019 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-31260753

RESUMEN

Mutations in the tumor protein (TP53) and the mammalian target of rapamycin (mTOR) pathway have been elucidated as driver mutations in ovarian carcinomas that transform into an invasive phenotype under hypoxic conditions. Chetomin (CHE) targets the hypoxic pathway while Everolimus (EVR) acts on the mTOR pathway. Poor aqueous solubilities of both compounds limit their clinical applications. Diblock copolymer nanoplatforms of methoxy poly(ethylene glycol)2000-block-poly (lactic acid)1800 (mPEG2000-b-PLA1800) and (mPEG4000-b-PLA2200) were used to formulate individual and dual drug loaded micelles (DDM) using the solvent evaporation method. The CHE micelles (CHE-M) had a size of 21 nm with CHE loading of 0.5 mg/mL while the EVR micelles (EVR-M) and the DDM had a size around 35 and 39 nm, respectively, with EVR loading up to 2.3 mg/mL. The anti-proliferative effects of these micelles have been tested in vitro in three ovarian cell lines (ES2, OVCAR3 and TOV21G) with the DDM exhibiting a strong synergistic anti-proliferative effect in the ES2 and the TOV21G cells. The DDM were able to significantly induce tumor regression in ES2 ovarian xenograft mouse models by inhibiting angiogenesis and inducing apoptosis when compared to the individual micelles. The inhibition of hypoxia inducible factor (HIF) and the mTOR pathways has been elucidated using immunohistochemistry studies. In conclusion, we have developed a mPEG-b-PLA based micellar nanoplatform that could prevent drug resistance by delivering multiple drugs at therapeutically relevant concentrations for effectively treating ovarian carcinomas.


Asunto(s)
Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Disulfuros/administración & dosificación , Portadores de Fármacos/administración & dosificación , Everolimus/administración & dosificación , Alcaloides Indólicos/administración & dosificación , Micelas , Neoplasias Ováricas/tratamiento farmacológico , Animales , Antineoplásicos/química , Protocolos de Quimioterapia Combinada Antineoplásica/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Disulfuros/química , Portadores de Fármacos/química , Combinación de Medicamentos , Liberación de Fármacos , Sinergismo Farmacológico , Everolimus/química , Femenino , Humanos , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Alcaloides Indólicos/química , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Poliésteres/administración & dosificación , Poliésteres/química , Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA