Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Mol Pharm ; 19(11): 4357-4369, 2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36282296

RESUMEN

CpG oligodeoxynucleotides are toll-like receptor 9 agonists capable of inducing potent pro-inflammatory immune responses. Although CpG oligodeoxynucleotides have shown promising antitumor effects, their systemic activity can trigger immune-related toxicity, limiting therapeutic application. We previously identified glatiramer acetate (GA), a cationic polypeptide approved for the treatment of relapsing-remitting multiple sclerosis, as an intratumoral delivery agent capable of complexing with CpG, thereby pinning it to the injection site and limiting systemic exposure. Here, we investigated whether the combination of CpG or GA-CpG polyplexes and intraperitoneal anti-PD-1 therapy would result in synergistic efficacy in AT84 and CT26 murine syngeneic models of head and neck and colon cancers, respectively. In both AT84 and CT26 tumor models, intratumoral CpG or GA-CpG treatment similarly suppressed tumor growth, but the efficacy was not amplified with anti-PD-1. Nevertheless, combination treatment increased cytotoxic T cell, helper T cell, and natural killer cell infiltration into AT84 tumors. Surprisingly, the combination of intratumoral GA and intraperitoneal anti-PD-1 treatment resulted in elevated systemic GM-CSF and IL-2 cytokine levels and demonstrated synergistic antitumor effects in the CT26 mouse tumor model. Moreover, tumors that responded most significantly to anti-PD-1 plus GA treatment showed increased markers of infiltration of CD4+ T cells and natural killer cells. Combinations of intratumoral GA or GA-CpG polyplexes with anti-PD-1 treatment warrant further investigation as combination cancer immunotherapy strategies.


Asunto(s)
Inmunoterapia , Neoplasias , Ratones , Animales , Acetato de Glatiramer/uso terapéutico , Inmunoterapia/métodos , Oligodesoxirribonucleótidos , Adyuvantes Inmunológicos/uso terapéutico , Adyuvantes Inmunológicos/farmacología , Neoplasias/tratamiento farmacológico , Línea Celular Tumoral
2.
Photodiagnosis Photodyn Ther ; 37: 102706, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34954388

RESUMEN

PURPOSE: Conventional photosensitizers for photodynamic therapy (PDT) typically have wide tissue distribution and poor water solubility. A hyaluronic acid (HA) polymeric nanoparticle with specific lymphatic uptake and highly water solubility was developed to deliver pyropheophorbide-a (PPa) for locally advanced head and neck squamous cell carcinoma (HNSCC) treatment. METHODS AND RESULTS: PPa was chemically conjugated to the HA polymeric nanoparticle via an adipic acid dihydrazide (ADH) linker. The conjugates were injected subcutaneously in a region near the tumor. Near-infrared (NIR) imaging was used to monitor distribution, and diode laser was used to activate PPa. The singlet oxygen generation efficiency of PPa was not affected by conjugation to HA nanoparticles at a PPa loading degree of 1.89 w.t.%. HA-ADH-PPa inhibited human HNSCC MDA-1986 cell growth only when photo-irradiation was applied. After HA-ADH-PPa treatment and radiation, NU/NU mice bearing human HNSCC MDA-1986 tumors showed reduced tumor growth and significantly enhanced survival time compared with an untreated group (p < 0.05). CONCLUSIONS: These results demonstrate that HA-ADH-PPa could be useful for in vivo locoregional photodynamic therapy of HNSCC.


Asunto(s)
Neoplasias de Cabeza y Cuello , Nanopartículas , Fotoquimioterapia , Animales , Línea Celular Tumoral , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Ácido Hialurónico/farmacología , Ratones , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico
3.
J Control Release ; 336: 410-432, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34171445

RESUMEN

With the significant drawbacks of conventional cancer chemotherapeutics, cancer immunotherapy has demonstrated the ability to eradicate cancer cells and circumvent multidrug resistance (MDR) with fewer side effects than traditional cytotoxic therapies. Various immunotherapeutic agents have been investigated for that purpose including checkpoint inhibitors, cytokines, monoclonal antibodies and cancer vaccines. All these agents aid immune cells to recognize and engage tumor cells by acting on tumor-specific pathways, antigens or cellular targets. However, immunotherapeutics are still associated with some concerns such as off-target side effects and poor pharmacokinetics. Nanomedicine may resolve some limitations of current immunotherapeutics such as localizing delivery, controlling release and enhancing the pharmacokinetic profile. Herein, we discuss recent advances of immunotherapeutic agents with respect to their development and biological mechanisms of action, along with the advantages that nanomedicine strategies lend to immunotherapeutics by possibly improving therapeutic outcomes and minimizing side effects.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Biología , Humanos , Inmunoterapia , Nanomedicina , Neoplasias/terapia
4.
Macromol Biosci ; 20(12): e2000251, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32924274

RESUMEN

Designing an in vitro model of the tumor extracellular microenvironment to screen intratumoral drugs is an active challenge. As recent clinical successes of human intratumoral therapies stimulate research on intratumoral delivery, a need for a 3D tumor model to screen intratumoral therapies arises. When injecting the drug formulation directly into the tumor, the biophysics affecting intratumoral retention must be considered; especially for biologic therapies, which may be dominated by extracellular transport mechanisms. Fibrotic regions in solid tumors are typically rich in collagen I fibers. Using shear rheology, head and neck tumors with higher collagen density show a higher stiffness. Similarly, the stiffness of the hyaluronic acid (HA) hydrogel models is increased by adding collagen fibers to model the bulk biomechanical properties of solid tumors. HA hydrogels are then used as intratumoral injection site simulators to model in vitro the retention of glatiramer acetate (GA) and polyethylene glycol (PEG) administered intratumorally. Both compounds are also injected in murine tumors and retention is studied ex vivo for comparison. Retention of GA in the hydrogels is significantly longer than PEG, analogous to the solid tumors, suggesting the utility of HA hydrogels with collagen I fibers for screening extracellular drug transport after intratumoral administration.


Asunto(s)
Materiales Biocompatibles/farmacología , Sistemas de Liberación de Medicamentos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Hidrogeles/farmacología , Animales , Materiales Biocompatibles/metabolismo , Línea Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Composición de Medicamentos , Acetato de Glatiramer/química , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Ácido Hialurónico/química , Hidrogeles/química , Ratones , Polietilenglicoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Control Release ; 326: 203-221, 2020 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-32673633

RESUMEN

Cancer therapies aim to kill tumor cells directly or engage the immune system to fight malignancy. Checkpoint inhibitors, oncolytic viruses, cell-based immunotherapies, cytokines, and adjuvants have been applied to prompt the immune system to recognize and attack cancer cells. However, systemic exposure of cancer therapies can induce unwanted adverse events. Intratumoral administration of potent therapies utilizes small amounts of drugs, in an effort to minimize systemic exposure and off-target toxicities. Here, we discuss the properties of the tumor microenvironment and transport considerations for intratumoral drug delivery. Specifically, we consider various tumor tissue factors and physicochemical factors that can affect tumor retention after intratumoral injection. We also review approved and clinical-stage intratumoral therapies and consider how the molecular and biophysical properties (e.g. size and charge) of these therapies influences intratumoral transport (e.g. tumor retention and cellular uptake). Finally, we offer a critical review and highlight several emerging approaches to promote tumor retention and limit systemic exposure of potent intratumoral therapies.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Preparaciones Farmacéuticas , Humanos , Inmunoterapia , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
6.
Bioorg Med Chem Lett ; 30(2): 126788, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31784317

RESUMEN

Small molecule agonists of TLR7/8, such as imidazoquinolines, are validated agonists for the treatment of cancer and for use in vaccine adjuvants. Imidazoquinolines have been extensively modified to understand the structure-activity relationship (SAR) at the N1- and C2-positions resulting in the clinical drug imiquimod, resiquimod, and several other highly potent analogues. However, the SAR of the aryl ring has not been fully elucidated in the literature. This initial study examines the SAR of C7-substituted imidazoquinolines. These compounds not only demonstrated that TLR7/8 tolerate changes at the C7-position but can increase potency and change their cytokine profiles. The most notable TLR7/8 agonists developed from this study 5, 8, and 14 which are up to 4-fold and 2-fold more active than resiquimod for TLR8 and/or TLR7, respectively, and up to 100-fold more active than the FDA approved imiquimod for TLR7.


Asunto(s)
Quinolinas/química , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Sitios de Unión , Citocinas/metabolismo , Humanos , Imidazoles/química , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Simulación del Acoplamiento Molecular , Quinolinas/metabolismo , Relación Estructura-Actividad , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo
7.
J Pharm Sci ; 108(10): 3372-3381, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31216451

RESUMEN

Oxidation of therapeutic proteins (TPs) can lead to changes in their pharmacokinetics, biological activity and immunogenicity. Metal impurities such as iron are known to increase oxidation of TPs, but nanoparticulate metals have unique physical and chemical properties compared to the bulk material or free metal ions. Iron oxide nanoparticles (IONPs) may originate from equipment used in the manufacturing of TPs or from needles during injection. In this study, the impact of IONPs on oxidation of a model protein, rat growth hormone (rGH), was investigated under chemical stress. Hydrogen peroxide (H2O2)- and 2,2'-azobis (2-methylpropionamidine) dihydrochloride oxidized methionine residues of rGH, but unexpectedly, oxidation was suppressed in the presence of IONPs compared to a phosphate buffer control. Fourier transform infrared spectroscopy indicated splitting of the α-helical absorbance band in the presence of IONPs, whereas circular dichroism spectra showed a reduced α-helical contribution with increasing temperature for both rGH and rGH-IONP mixtures. The results collectively indicate that IONPs can increase the chemical stability of rGH by altering the kinetics and preference of amino acid residues that are oxidized, although the changes in protein secondary structure by IONPs may lead to alterations of physical stability.


Asunto(s)
Compuestos Férricos/química , Hormona del Crecimiento/química , Hierro/química , Nanopartículas/química , Oxidación-Reducción/efectos de los fármacos , Estructura Secundaria de Proteína/efectos de los fármacos , Aminoácidos/química , Animales , Dicroismo Circular , Peróxido de Hidrógeno/química , Conformación Proteica en Hélice alfa/efectos de los fármacos , Ratas
8.
J Control Release ; 306: 165-176, 2019 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-31173789

RESUMEN

The toll-like receptor 7 and 8 (TLR7/8) agonist Resiquimod (R848) has been recognized as a promising immunostimulator for the treatment of cutaneous cancers in multiple clinical trials. However, systemic administration of R848 often results in strong immune-related toxicities while having limited therapeutic effects to the tumor. In the present study, a prodrug-based nanocarrier delivery system was developed that exhibited high therapeutic efficiency. R848 was conjugated to α-tocopherol to constitute an R848-Toco prodrug, followed by formulating with a tocopherol-modified hyaluronic acid (HA-Toco) as a polymeric nano-suspension. In vitro evaluation showed that the delivery system prolonged the release kinetics while maintaining TLR agonist activities. When administered subcutaneously, the nano-suspension formed a depot at the injection site, inducing localized immune responses without systemic expansion. This formulation also suppressed tumor growth and recruited immune cells to the tumor in a murine model of head and neck cancer. In a preclinical canine study of spontaneous mast cell tumors, the treatment led to a 67% response rate (three partial remissions and one complete remission).


Asunto(s)
Antineoplásicos/farmacología , Imidazoles/administración & dosificación , Factores Inmunológicos/farmacología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Animales , Citocinas/metabolismo , Perros , Composición de Medicamentos , Evaluación Preclínica de Medicamentos , Liberación de Fármacos , Humanos , Imidazoles/química , Imidazoles/farmacología , Ratones , Ratones Endogámicos C3H , Conejos , Suspensiones
9.
Pharm Res ; 35(8): 162, 2018 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-29931580

RESUMEN

PURPOSE: To investigate influence of inflammation on metabolism and pharmacokinetics (PK) of midazolam (MDZ) and construct a semi-physiologically based pharmacokinetic (PBPK) model to predict PK in mice with inflammatory disease. METHODS: Glucose-6-phosphate isomerase (GPI)-mediated inflammation was used as a preclinical model of arthritis in DBA/1 mice. CYP3A substrate MDZ was selected to study changes in metabolism and PK during the inflammation. The semi-PBPK model was constructed using mouse physiological parameters, liver microsome metabolism, and healthy animal PK data. In addition, serum cytokine, and liver-CYP (cytochrome P450 enzymes) mRNA levels were examined. RESULTS: The in vitro metabolite formation rate was suppressed in liver microsomes prepared from the GPI-treated mice as compared to the healthy mice. Further, clearance of MDZ was reduced during inflammation as compared to the healthy group. Finally, the semi-PBPK model was used to predict PK of MDZ after GPI-mediated inflammation. IL-6 and TNF-α levels were elevated and liver-cyp3a11 mRNA was reduced after GPI treatment. CONCLUSION: The semi-PBPK model successfully predicted PK parameters of MDZ in the disease state. The model may be applied to predict PK of other drugs under disease conditions using healthy animal PK and liver microsomal data as inputs.


Asunto(s)
Adyuvantes Anestésicos/farmacocinética , Inflamación/metabolismo , Midazolam/farmacocinética , Adyuvantes Anestésicos/metabolismo , Animales , Citocromo P-450 CYP3A/metabolismo , Glucosa-6-Fosfato Isomerasa/metabolismo , Humanos , Masculino , Ratones , Microsomas Hepáticos/metabolismo , Midazolam/metabolismo , Modelos Biológicos
10.
Mol Cancer Res ; 16(6): 961-973, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29545475

RESUMEN

FOXM1 transcription factor network is activated in over 84% of cases in high-grade serous ovarian cancer (HGSOC), and FOXM1 upregulates the expression of genes involved in the homologous recombination (HR) DNA damage and repair (DDR) pathway. However, the role of FOXM1 in PARP inhibitor response has not yet been studied. This study demonstrates that PARP inhibitor (PARPi), olaparib, induces the expression and nuclear localization of FOXM1. On the basis of ChIP-qPCR, olaparib enhances the binding of FOXM1 to genes involved in HR repair. FOXM1 knockdown by RNAi or inhibition by thiostrepton decreases FOXM1 expression, decreases the expression of HR repair genes, such as BRCA1 and RAD51, and enhances sensitivity to olaparib. Comet and PARP trapping assays revealed increases in DNA damage and PARP trapping in FOXM1-inhibited cells treated with olaparib. Finally, thiostrepton decreases the expression of BRCA1 in rucaparib-resistant cells and enhances sensitivity to rucaparib. Collectively, these results identify that FOXM1 plays an important role in the adaptive response induced by olaparib and FOXM1 inhibition by thiostrepton induces "BRCAness" and enhances sensitivity to PARP inhibitors.Implications: FOXM1 inhibition represents an effective strategy to overcome resistance to PARPi, and targeting FOXM1-mediated adaptive pathways may produce better therapeutic effects for PARP inhibitors. Mol Cancer Res; 16(6); 961-73. ©2018 AACR.


Asunto(s)
Antineoplásicos/efectos adversos , Proteína Forkhead Box M1/genética , Ftalazinas/efectos adversos , Piperazinas/efectos adversos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Ftalazinas/farmacología , Piperazinas/farmacología , Transfección
11.
Pharmaceutics ; 9(3)2017 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-28902176

RESUMEN

Doxorubicin (Dox) is an effective anti-cancer medication with poor oral bioavailability and systemic toxicities. DoxQ was developed by conjugating Dox to the lymphatically absorbed antioxidant quercetin to improve Dox's bioavailability and tolerability. The purpose of this study was to characterize the pharmacokinetics and safety of Dox after intravenous (IV) and oral (PO) administration of DoxQ or Dox (10 mg/kg) and investigate the intestinal lymphatic delivery of Dox after PO DoxQ administration in male Sprague-Dawley rats. Drug concentrations in serum, urine, and lymph were quantified by HPLC with fluorescence detection. DoxQ intact IV showed a 5-fold increase in the area under the curve (AUC)-18.6 ± 1.98 compared to 3.97 ± 0.71 µg * h/mL after Dox-and a significant reduction in the volume of distribution (Vss): 0.138 ± 0.015 versus 6.35 ± 1.06 L/kg. The fraction excreted unchanged in urine (fe) of IV DoxQ and Dox was ~5% and ~11%, respectively. Cumulative amounts of Dox in the mesenteric lymph fluid after oral DoxQ were twice as high as Dox in a mesenteric lymph duct cannulation rat model. Oral DoxQ increased AUC of Dox by ~1.5-fold compared to after oral Dox. Concentrations of ß-N-Acetylglucosaminidase (NAG) but not cardiac troponin (cTnI) were lower after IV DoxQ than Dox. DoxQ altered the pharmacokinetic disposition of Dox, improved its renal safety and oral bioavailability, and is in part transported through intestinal lymphatics.

12.
Eur J Med Chem ; 136: 452-456, 2017 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-28525843

RESUMEN

A new pH-activated polymer chelate of cisplatin was synthesized using a scalable and green aqueous technique. Synthesis of the chelate was based on formation of a 6-member ring of platinum(II) with acetyl-homo-Lysine (Ac-homo-Lys), which was accomplished under completely aqueous conditions using a traceless photocleavable protection chemistry. Synthesis preceded by, first, amidation of a photocaged homo-Ac-Lys with hyaluronic acid (HA) in water using a p-hydroxyphenacyl (pHP) group as the photoremovable protecting group, followed by reaction of cisplatin (diaqua form) in water to form the reversible chelate. Platinum drug release was pH rate controlled, with more rapid release (t1/2 20 h) at acidic pH similar to the tumor microenvironment yet slower release (t1/2 35 h) at normal physiological pH.


Asunto(s)
Antineoplásicos/química , Quelantes/química , Sistemas de Liberación de Medicamentos , Compuestos Organoplatinos/química , Polímeros/química , Antineoplásicos/síntesis química , Quelantes/síntesis química , Portadores de Fármacos/síntesis química , Portadores de Fármacos/química , Humanos , Concentración de Iones de Hidrógeno , Modelos Moleculares , Estructura Molecular , Compuestos Organoplatinos/síntesis química , Polímeros/síntesis química
13.
Drug Deliv Transl Res ; 7(4): 582-597, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28462502

RESUMEN

Doxorubicin is an effective anticancer drug; however, it is cardiotoxic and has poor oral bioavazilability. Quercetin is a plant-based flavonoid with inhibitory effects on P-glycoprotein (P-gp) and CYP3A4 and also antioxidant properties. To mitigate these therapeutic barriers, DoxQ, a novel derivative of doxorubicin, was synthesized by conjugating quercetin to doxorubicin. The purpose of this study is to mechanistically elucidate the in vitro safety and efficacy of DoxQ. Drug release in vitro and cellular uptake by multidrug-resistant canine kidney (MDCK-MDR) cells were quantified by HPLC. Antioxidant activity, CYP3A4 inhibition, and P-gp inhibitory effects were examined using commercial assay kits. Drug potency was assessed utilizing triple-negative murine breast cancer cells, and cardiotoxicity was assessed utilizing adult rat and human cardiomyocytes (RL-14). Levels of reactive oxygen species and gene expression of cardiotoxicity markers, oxidative stress markers, and CYP1B1 were determined in RL-14. DoxQ was less cytotoxic to both rat and human cardiomyocytes and retained anticancer activity. Levels of ROS and markers of oxidative stress demonstrate lower oxidative damage induced by DoxQ compared to doxorubicin. DoxQ also inhibited the expression and catalytic activity of CYP1B1. Additionally, DoxQ inhibited CYP3A4 and demonstrated higher cellular uptake by MDCK-MDR cells than doxorubicin. DoxQ provides a novel therapeutic approach to mitigate the cardiotoxicity and poor oral bioavailability of doxorubicin. The cardioprotective mechanism of DoxQ likely involves scavenging ROS and CYP1B1 inhibition, while the mechanism of improving the poor oral bioavailability of doxorubicin is likely related to inhibiting CYP3A4 and P-gp.


Asunto(s)
Antineoplásicos/farmacología , Antioxidantes/farmacología , Inhibidores del Citocromo P-450 CYP3A/farmacología , Doxorrubicina/farmacología , Quercetina/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Antineoplásicos/química , Antioxidantes/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citocromo P-450 CYP1B1/genética , Citocromo P-450 CYP1B1/metabolismo , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A/química , Perros , Doxorrubicina/química , Liberación de Fármacos , Humanos , Células de Riñón Canino Madin Darby , Miocitos Cardíacos/efectos de los fármacos , Quercetina/química , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transcitosis
14.
Am J Vet Res ; 77(9): 1005-16, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27580113

RESUMEN

OBJECTIVE To conduct a phase I-II clinical trial of hyaluronan-cisplatin nanoconjugate (HA-Pt) in dogs with naturally occurring malignant tumors. ANIMALS 18 healthy rats, 9 healthy mice, and 16 dogs with cancer. PROCEDURES HA-Pt was prepared and tested by inductively coupled plasma mass spectrometry; DNA-platinum adduct formation and antiproliferation effects of cisplatin and HA-Pt were compared in vitro. Effects of cisplatin (IV) and HA-Pt (SC) in rodents were tested by clinicopathologic assays. In the clinical trial, dogs with cancer received 1 to 4 injections of HA-Pt (10 to 30 mg/m(2), intratumoral or peritumoral, q 3 wk). Blood samples were collected for pharmacokinetic analysis; CBC, serum BUN and creatinine concentration measurement, and urinalysis were conducted before and 1 week after each treatment. Some dogs underwent hepatic enzyme testing. Tumors were measured before the first treatment and 3 weeks after each treatment to assess response. RESULTS No adverse drug effects were detected in pretrial assessments in rodents. Seven of 16 dogs completed the study; 3 had complete tumor responses, 3 had stable disease, and 1 had progressive disease. Three of 7 dogs with oral and nasal squamous cell carcinoma (SCC) that completed the study had complete responses. Myelosuppression and cardiotoxicosis were identified in 6 and 2 dogs, respectively; none had nephrotoxicosis. Four of 5 dogs with hepatic enzymes assessed had increased ALT activities, attributed to diaquated cisplatin products in the HA-Pt. Pharmacokinetic data fit a 3-compartment model. CONCLUSIONS AND CLINICAL RELEVANCE HA-Pt treatment resulted in positive tumor responses in some dogs, primarily those with SCC. The adverse effect rate was high. IMPACT FOR HUMAN MEDICINE Oral SCC in dogs has characteristics similar to human head and neck SCC; these results could be useful in developing human treatments.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Enfermedades de los Perros/tratamiento farmacológico , Ácido Hialurónico/uso terapéutico , Nanoconjugados/uso terapéutico , Neoplasias/veterinaria , Animales , Antineoplásicos/farmacocinética , Cisplatino/farmacocinética , Perros , Femenino , Ácido Hialurónico/farmacocinética , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Inducción de Remisión
15.
Appl Spectrosc ; 70(9): 1529-36, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27527103

RESUMEN

Cisplatin, a platinum chemotherapeutic, is one of the most commonly used chemotherapeutic agents for many solid tumors. In this work, we developed and validated an inductively coupled plasma mass spectrometry (ICP-MS) method for quantitative determination of platinum levels in rat urine, plasma, and tissue matrices including liver, brain, lungs, kidney, muscle, heart, spleen, bladder, and lymph nodes. The tissues were processed using a microwave accelerated reaction system (MARS) system prior to analysis on an Agilent 7500 ICP-MS. According to the Food and Drug Administration guidance for industry, bioanalytical validation parameters of the method, such as selectivity, accuracy, precision, recovery, and stability were evaluated in rat biological samples. Our data suggested that the method was selective for platinum without interferences caused by other presenting elements, and the lower limit of quantification was 0.5 ppb. The accuracy and precision of the method were within 15% variation and the recoveries of platinum for all tissue matrices examined were determined to be 85-115% of the theoretical values. The stability of the platinum-containing solutions, including calibration standards, stock solutions, and processed samples in rat biological matrices was investigated. Results indicated that the samples were stable after three cycles of freeze-thaw and for up to three months.


Asunto(s)
Espectrometría de Masas/métodos , Platino (Metal)/análisis , Platino (Metal)/farmacocinética , Animales , Estabilidad de Medicamentos , Modelos Lineales , Platino (Metal)/química , Ratas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Distribución Tisular
16.
Pharm Res ; 33(10): 2517-29, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27335023

RESUMEN

PURPOSE: To determine aerosol administration capability and therapeutic efficacy of the new formulation of hyaluronan cisplatin conjugates, HylaPlat™ (HA-Pt), for lung cancer treatment. METHODS: In vitro formulation stability test, 2D and 3D spheroid cell culture and in vivo efficacy studies using mouse orthotopic allograft models were conducted. RESULTS: The HA-Pt effectively attenuated cell growth in 2D and 3D cultures with IC50 of 2.62 and 5.36 µM, respectively, which were comparable to those with unconjugated control cisplatin-dependent growth inhibition (IC50 1.64 and 4.63 µM, respectively). A single dose of either 7.5 or 15 mg/kg HA-Pt (cisplatin equivalent) by intratracheal aerosol spray 7 days after Lewis lung carcinoma (LLC) cell inoculation markedly inhibited growth of LLC allografts in mouse lungs and resulted in a 90 or 94% reduction of tumor nodule numbers, respectively, as compared to those from the PBS control. Cancer stem cells and cisplatin resistant cells marker, CD44 expression decreased in the tumor nodules of the HA-Pt but not in those of cisplatin treated groups. CONCLUSIONS: The current study suggests that an intratracheal aerosol administration of the HA-Pt nanoparticles offers an effective strategy for lung cancer treatment and this treatment may induce only limited cisplatin resistance.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Ácido Hialurónico/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Nanopartículas/administración & dosificación , Tráquea/metabolismo , Células A549 , Administración por Inhalación , Animales , Antineoplásicos/metabolismo , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Cisplatino/metabolismo , Vías de Administración de Medicamentos , Femenino , Humanos , Ácido Hialurónico/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos C57BL , Nanopartículas/metabolismo , Tráquea/efectos de los fármacos , Células Tumorales Cultivadas
17.
J Pharm Sci ; 105(6): 1891-1900, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27155765

RESUMEN

The purpose of this study was to develop a safe and efficacious drug delivery platform for sustained release of cisplatin after locoregional administration. We successfully synthesized hyaluronan-cisplatin nanoconjugates (HA-Lys-Pt) using an N-Ac-lysine linker, which formed a thermodynamically stable five-membered ring with the platinum. The conjugate was characterized for release kinetics, in vitro anti-proliferative activity, degradability, impurity content, formation of Pt-DNA adducts, pharmacokinetics, tolerability in rodents and canines, and for efficacy in rodents. The 75 kD HA-Lys-Pt (75HA-Lys-Pt) sustained release of platinum with a 69 h half-life in phosphate buffered saline without substantial burst release. Compared to intravenous cisplatin, subcutaneously injected 75HA-Lys-Pt formed 3.2-fold more Pt-DNA adducts in rat peripheral blood mononuclear cells compared to intravenous cisplatin over 96 h. Subcutaneous 75HA-Lys-Pt was tolerable in rats at 40 mg/kg (4 × LD50 of conventional cisplatin) and resulted in 62.5% partial response and 37.5% stable disease in murine xenografts of head and neck squamous cell cancer (20 mg/kg/wk × 3 weeks). 75HA-Lys-Pt demonstrated extended tmax and improved area-under-the-curve compared to cisplatin in rats and canines. Canine safety was demonstrated by liver enzyme and electrolyte levels, complete blood count, and urinalysis.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Portadores de Fármacos/administración & dosificación , Ácido Hialurónico/administración & dosificación , Lisina/administración & dosificación , Neoplasias/metabolismo , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Cisplatino/farmacocinética , Perros , Relación Dosis-Respuesta a Droga , Portadores de Fármacos/farmacocinética , Femenino , Humanos , Ácido Hialurónico/farmacocinética , Lisina/farmacocinética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Proyectos Piloto , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
18.
Laryngoscope ; 126(5): E184-90, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26690734

RESUMEN

OBJECTIVES/HYPOTHESIS: To evaluate the efficacy of peritumoral hyaluronic acid (HA)-cisplatin therapy in a murine model of laryngeal squamous cell carcinoma and to evaluate its effect on cancer stem cells (CSCs). STUDY DESIGN: An orthotopic murine study utilizing University of Michigan squamous cell carcinoma-12 (UMSCC-12) laryngeal cancer cells was conducted in randomized controlled fashion with three treatment arms: saline, systemic cisplatin, and peritumoral HA-cisplatin. METHODS: UMSCC-12 laryngeal cancer cells were inoculated into the buccal mucosa of athymic nude mice followed by weekly treatment with saline, systemic cisplatin, or peritumoral HA-cisplatin for 3 weeks. Tumor response and animal weight was monitored and change in CD44 proportion was analyzed ex vivo. RESULTS: HA-cisplatin demonstrated superior antitumor efficacy and greater reduction in CD44 positivity on ex vivo analysis. CONCLUSIONS: Peritumoral nanoconjugated HA-cisplatin provides superior antitumor efficacy compared to standard cisplatin therapy in an in vivo laryngeal cancer model. There was also selective targeting of CD44+ cancer cells with HA-cisplatin. This therapeutic strategy could represent the first selective laryngeal CSC-targeted therapy. Further preclinical investigation is warranted to evaluate its role for locally advanced head and neck cancer treatment. LEVEL OF EVIDENCE: NA Laryngoscope, 126:E184-E190, 2016.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Cisplatino/farmacología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias Laríngeas/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Adyuvantes Inmunológicos/farmacología , Animales , Humanos , Receptores de Hialuranos/efectos de los fármacos , Ácido Hialurónico/farmacología , Ratones , Ratones Desnudos , Nanoconjugados/administración & dosificación , Carcinoma de Células Escamosas de Cabeza y Cuello
19.
Ther Deliv ; 6(10): 1195-210, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26606855

RESUMEN

BACKGROUND: A biocompatible core/shell structured magnetic nanoparticles (MNPs) was developed to mediate simultaneous cancer therapy and imaging. METHODS & RESULTS: A 22-nm MNP was first synthesized via magnetically coupling hard (FePt) and soft (Fe3O4) materials to produce high relative energy transfer. Colloidal stability of the FePt@Fe3O4 MNPs was achieved through surface modification with silane-polyethylene glycol (PEG). Intravenous administration of PEG-MNPs into tumor-bearing mice resulted in a sustained particle accumulation in the tumor region, and the tumor burden of treated mice was a third that of the mice in control groups 2 weeks after a local hyperthermia treatment. In vivo magnetic resonance imaging exhibited enhanced T2 contrast in the tumor region. CONCLUSION: This work has demonstrated the feasibility of cancer theranostics with PEG-MNPs.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Hipotermia Inducida/métodos , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita/administración & dosificación , Polietilenglicoles/administración & dosificación , Administración Intravenosa , Animales , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Medios de Contraste/administración & dosificación , Femenino , Ratones , Ratones Endogámicos BALB C , Propiedades de Superficie
20.
Tissue Eng Part A ; 21(11-12): 1795-809, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25760435

RESUMEN

The transcription factor atonal homolog 1 (ATOH1) has multiple homologues that are functionally conserved across species and is responsible for the generation of sensory hair cells. To evaluate potential functional differences between homologues, human and mouse ATOH1 (HATH1 and MATH-1, respectively) were nonvirally delivered to human Wharton's jelly cells (hWJCs) for the first time. Delivery of HATH1 to hWJCs demonstrated superior expression of inner ear hair cell markers and characteristics than delivery of MATH-1. Inhibition of HES1 and HES5 signaling further increased the atonal effect. Transfection of hWJCs with HATH1 DNA, HES1 siRNA, and HES5 siRNA displayed positive identification of key hair cell and support cell markers found in the cochlea, as well as a variety of cell shapes, sizes, and features not native to hair cells, suggesting the need for further examination of other cell types induced by HATH1 expression. In the first side-by-side evaluation of HATH1 and MATH-1 in human cells, substantial differences were observed, suggesting that the two atonal homologues may not be interchangeable in human cells, and artificial expression of HATH1 in hWJCs requires further study. In the future, this line of research may lead to engineered systems that would allow for evaluation of drug ototoxicity or potentially even direct therapeutic use.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Técnicas de Reprogramación Celular/métodos , Células Ciliadas Auditivas Internas/citología , Células Madre Mesenquimatosas/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Linaje de la Célula , Femenino , Colorantes Fluorescentes/metabolismo , Vectores Genéticos/genética , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Humanos , Recién Nacido , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Miosina VIIa , Miosinas/biosíntesis , Miosinas/genética , Compuestos de Piridinio/metabolismo , Compuestos de Amonio Cuaternario/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Transducción de Señal , Especificidad de la Especie , Factor de Transcripción HES-1 , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA