Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Molecules ; 27(11)2022 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-35684309

RESUMEN

The anticancer agent doxorubicin(dox) has been widely used in the treatment of a variety of hematological malignancies and solid tumors. Despite doxorubicin's efficiency in killing tumor cells, severe damage to healthy tissues, along with cardiotoxicity, limits its clinical use. To overcome these adverse side effects, improve patient safety, and enhance therapeutic efficacy, we have designed a thermally responsive biopolymer doxorubicin carrier that can be specifically targeted to tumor tissue by locally applying mild hyperthermia (41 °C). The developed drug vehicle is composed of the following: a cell penetrating peptide (SynB1) to promote tumor and cellular uptake; thermally responsive Elastin-like polypeptide (ELP); and the (6-maleimidocaproyl) hydrazone derivative of doxorubicin (DOXO-EMCH) containing a pH-sensitive hydrazone linker that releases doxorubicin in the acidic tumor environment. We used the in vivo imaging system, IVIS, to determine biodistribution of doxorubicin-delivered ELP in MDA-MB-231 xenografts in nude mice. Tumor bearing mice were treated with a single IV injection of 10 mg/kg doxorubicin equivalent dose with free doxorubicin, thermally responsive SynB1 ELP 1-DOXO, and a thermally nonresponsive control biopolymer, SynB1 ELP 2-DOXO. Following a 2 h treatment with hyperthermia, tumors showed a 2-fold higher uptake when treated with SynB1 ELP 1-DOXO compared to free doxorubicin. Accumulation of the thermally non-responsive control SynB1 ELP2 -DOXO was comparable to free doxorubicin, indicating that an increase in dox accumulation with ELP is due to aggregation in response to thermal targeting. Higher levels of SynB1 ELP1-DOXO and SynB1 ELP2 -DOXO with respect to free doxorubicin were observed in kidneys. Fluorescence intensity from hearts of animals treated with SynB1 ELP1-DOXO show a 5-fold decrease in accumulation of doxorubicin than the same dose of free doxorubicin. SynB1-ELP1-DOXO biopolymers demonstrated a 6-fold increase in tumor/heart ratio in comparison to free doxorubicin, indicating preferential accumulation of the drug in tumors. These results demonstrate that thermally targeted polymers are a promising therapy to enhance tumor targeting and uptake of anticancer drugs and to minimize free drug toxicity in healthy tissues, representing a great potential for clinical application.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Péptidos de Penetración Celular , Hipertermia Inducida , Animales , Neoplasias de la Mama/tratamiento farmacológico , Cardiotoxicidad/prevención & control , Péptidos de Penetración Celular/farmacología , Doxorrubicina , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Hidrazonas , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Desnudos , Distribución Tisular
2.
Int J Mol Sci ; 22(3)2021 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-33498762

RESUMEN

Elastin-like polypeptides (ELPs) undergo a characteristic phase transition in response to ambient temperature. Therefore, it has been be used as a thermosensitive vector for the delivery of chemotherapy agents since it can be used to target hyperthermic tumors. This novel strategy introduces unprecedented options for treating cancer with fewer concerns about side effects. In this study, the ELP system was further modified with an enzyme-cleavable linker in order to release drugs within tumors. This system consists of an ELP, a matrix metalloproteinase (MMP) substrate, a cell-penetrating peptide (CPP), and a 6-maleimidocaproyl amide derivative of doxorubicin (Dox). This strategy shows up to a 4-fold increase in cell penetration and results in more death in breast cancer cells compared to ELP-Dox. Even in doxorubicin-resistant cells (NCI/ADR and MES-SA/Dx5), ELP-released cell-penetrating doxorubicin demonstrated better membrane penetration, leading to at least twice the killing of resistant cells compared to ELP-Dox and free Dox. MMP-digested CPP-Dox showed better membrane penetration and induced more cancer cell death in vitro. This CPP-complexed Dox released from the ELP killed even Dox-resistant cells more efficiently than both free doxorubicin and non-cleaved ELP-CPP-Dox.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Resistencia a Antineoplásicos/efectos de los fármacos , Antibióticos Antineoplásicos/farmacocinética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Péptidos de Penetración Celular/farmacocinética , Doxorrubicina/farmacocinética , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Liberación de Fármacos , Elastina/química , Femenino , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Péptidos/química , Péptidos/metabolismo , Rodaminas/química , Rodaminas/farmacocinética
3.
Molecules ; 20(12): 21750-69, 2015 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-26690101

RESUMEN

The majority of anticancer drugs have poor aqueous solubility, produce adverse effects in healthy tissue, and thus impose major limitations on both clinical efficacy and therapeutic safety of cancer chemotherapy. To help circumvent problems associated with solubility, most cancer drugs are now formulated with co-solubilizers. However, these agents often also introduce severe side effects, thereby restricting effective treatment and patient quality of life. A promising approach to addressing problems in anticancer drug solubility and selectivity is their conjugation with polymeric carriers to form polymer-based prodrugs. These polymer-based prodrugs are macromolecular carriers, designed to increase the aqueous solubility of antitumor drugs, can enhance bioavailability. Additionally, polymer-based prodrugs approach exploits unique features of tumor physiology to passively facilitate intratumoral accumulation, and so improve chemodrug pharmacokinetics and pharmacological properties. This review introduces basic concepts of polymer-based prodrugs, provides an overview of currently emerging synthetic, natural, and genetically engineered polymers that now deliver anticancer drugs in preclinical or clinical trials, and highlights their major anticipated applications in anticancer therapies.


Asunto(s)
Antineoplásicos/administración & dosificación , Portadores de Fármacos/administración & dosificación , Neoplasias/tratamiento farmacológico , Polímeros/administración & dosificación , Profármacos/administración & dosificación , Animales , Antineoplásicos/farmacocinética , Portadores de Fármacos/farmacocinética , Humanos , Profármacos/farmacocinética , Solubilidad
4.
Biochem Biophys Rep ; 31: 101326, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36039113

RESUMEN

Human epidermal growth factor receptor isoform D (EGFR; isoform D) is a soluble protein from a 3 kb alternate mRNA transcript that arises from the human EGFR gene. Several studies have identified this circulating isoform of EGFR as a potential diagnostic biomarker for the detection of early stage of cancers. While the expression of the full-length EGFR (isoform A) is regulated by its cognate ligand, EGF, as well as by phorbol myristate acetate (PMA), no studies have examined the factors regulating the expression of EGFR isoform D. In this study, using breast cancer cell lines, we show that the HER receptor ligands, EGF and neuregulin (NRG-1ß), as well as the phorbol ester, PMA, can increase the expression of EGFR isoform D, as well as isoform A. Our results, based on measurement of mRNA levels, suggest that EGF induced expression of both isoform A and isoform D occur through a mitogen activated protein kinase (MAPK)-dependent mechanism, and also suggest that protein kinase C is involved in PMA-induced regulation of both isoforms. We also demonstrate that NRG-1ß increases isoform A and isoform D expression via the MAPK-dependent pathway, but this regulation occurs independently of phosphatidylinositol 3-kinase/Akt activation. These results suggest that regulation of EGFR isoform A and isoform D expression occur using similar mechanisms. Despite commonalities in the transcriptional regulation of these two EGFR isoforms, the half-lives of these two transcripts is quite different. Moreover, EGFR isoform D, unlike isoform A, is not post-transcriptionally modulated by EGFR activators in the breast cancer cell line MDA-MB-468.

5.
J Chemother ; 31(1): 23-29, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30773131

RESUMEN

This research describes a thermally responsive elastin-like polypeptide (ELP) for the delivery of dnMAML peptides that inhibit the Notch pathway. Exploiting passive targeting and a thermally active tumor-targeting technique available through the use of ELP, the dnMAML peptide was efficiently delivered to tumor tissue. Furthermore, this ELP-dnMAML was modified with the addition of a cell penetrating peptide (SynB1) for improved infiltration of ELP-dnMAML into the tumor cells. In this study, we verified that intravenously delivered SynB1-ELP-dnMAML was cleared from circulation under physiological conditions (37 °C) but accumulated at tumors grown in mice at sites to which an externally induced, local heat (40-41 °C) was applied, thereby resulting in greatly reduced tumor growth in animals. Additionally, in combination with Taxol, SynB1-ELP-dnMAML showed more potent tumor growth retardation.


Asunto(s)
Proteínas de Unión al ADN/administración & dosificación , Sistemas de Liberación de Medicamentos , Neoplasias Mamarias Experimentales/patología , Paclitaxel/administración & dosificación , Péptidos/administración & dosificación , Receptores Notch/antagonistas & inhibidores , Factores de Transcripción/administración & dosificación , Animales , Antineoplásicos Fitogénicos , Línea Celular Tumoral , Péptidos de Penetración Celular , Femenino , Humanos , Hipertermia Inducida , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Ther Deliv ; 10(4): 251-263, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30991923

RESUMEN

Therapeutic strategies for traumatic injuries in the central nervous system (CNS) are largely limited to the efficiency of drug delivery. Despite the disrupted blood-CNS barrier during the early phase after injury, the drug administration faces a variety of obstacles derived from homeostatic imbalance at the injury site. In the late phase after CNS injury, the restoration of the blood-CNS barrier integrity varies depending on the injury severity resulting in inconsistent delivery of therapeutics. This review intends to characterize those different challenges of the therapeutic delivery in acute and chronic phases after injury and discuss recent advances in various approaches to explore novel strategies for the treatment of traumatic CNS injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Enfermedad Aguda , Animales , Barrera Hematoencefálica/metabolismo , Lesiones Traumáticas del Encéfalo/fisiopatología , Enfermedad Crónica , Humanos , Traumatismos de la Médula Espinal/fisiopatología , Distribución Tisular , Índices de Gravedad del Trauma
7.
Arch Pharm Res ; 31(10): 1355-61, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18958428

RESUMEN

We investigated the anticancer activity of 11-hydroxyaclacinomycin X (ID-6105), a novel anthracycline, on weakly doxorubicin (Dox)-resistant SK-OV-3 ovarian cancer cells, and elucidated the relationship between its anticancer activity and accumulation in cells compared with those of Dox. Accumulation of ID-6105 in the cells was time-and concentration-dependent, a result of drug-induced cytotoxicity in the cells. SK-OV-3 cells were preloaded with ID-6105 or Dox for 12 h at concentrations ranging from 100 to 2000 nM and then incubated with drug-free medium for 0-48 h. Cell viability was measured using a proliferation-based assay (XTT assay). The inhibitory effects of ID-6105 on cell viability were more pronounced than those of Dox. The IC(50) values of ID-6105 after 24-and 48-h incubation with drug-free medium were 1.58 and 0.084 microM, while those of Dox were 2 and 0.334 microM, respectively. To investigate the relationship between the intracellular levels and the cytotoxic effects of the drugs, we preloaded SKOV-3 cells with ID-6105 or Dox (100-2000 nM) for 12 h and then measured the intracellular levels of drugs by HPLC in drug-free medium for 0-48 h. After preloading the drugs, the intracellular concentrations of ID-6105 at time 0 were 1.3-, 1.8-, and 1.4-fold larger than those of Dox at initial concentrations of 500, 1000, and 2000 nM, respectively. The extent of ID-6105 accumulation in the cells was more pronounced than that of Dox. These findings suggest that ID-6105 effluxed less from the cells than Dox, resulting in its extensive cytotoxicity compared with that of Dox. These results show that accumulation of ID-6105 within tumor cells may be important for the inhibitory effects of this drug in cancer cells. ID-6105 has an antiproliferative effect on SK-OV-3 cells that is due to its cytotoxicity. This effect is more pronounced than that of Dox, and may be attributed to extensive accumulation of ID-6105 in the cells.


Asunto(s)
Aclarubicina/análogos & derivados , Antineoplásicos/metabolismo , Neoplasias Ováricas/metabolismo , Aclarubicina/metabolismo , Aclarubicina/farmacología , Antibióticos Antineoplásicos/metabolismo , Antibióticos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Doxorrubicina/metabolismo , Doxorrubicina/farmacología , Femenino , Humanos , Indicadores y Reactivos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología
8.
Cancer Chemother Pharmacol ; 60(2): 197-201, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17096162

RESUMEN

A new anthracycline ID6105 (11-hydroxyaclacinomycin X, Hyrubicin), which has potent antitumor activities against a broad range of cancer cell lines, was produced by hybrid biosynthetic approach. We investigated ID6105-induced apoptosis and in vivo efficacy on experimental tumors, and also defined its optimal dosing schedule. From PARP cleavage assay and caspase-3 activation assay, we found that ID6105 can induce apoptosis in tumor cells and its ability was superior to doxorubicin. In human tumor xenograft models, ID6105 showed greater antitumor effects on SW620 and NCI-H23 than doxorubicin. The 1 mg/kg of ID6105 treatment reduced size of tumor by 93% in NCI-H23 model whereas doxorubicin (2 mg/kg) showed only 39% inhibition rate. In SW620 model, 0.3 mg/kg of ID6105 proved to be comparable to 2 mg/kg of doxorubicin. Testing with several dosing schedule such as qd10, qd5, and q4d3, we decided intravenous qd5 treatment was an optimal schedule as a dose regimen of ID6105. In conclusion, ID6105 is a potent apoptosis-inducing anthracycline and effective in treatment of tumors with qd5 dosing schedule.


Asunto(s)
Aclarubicina/análogos & derivados , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Neoplasias Experimentales/tratamiento farmacológico , Aclarubicina/uso terapéutico , Animales , Caspasa 3/metabolismo , Doxorrubicina/farmacología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Experimentales/patología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Organismos Libres de Patógenos Específicos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Arch Pharm Res ; 30(11): 1419-25, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18087810

RESUMEN

We investigated the effects of hydrogen peroxide (H2O2) on relaxation of the cat lower esophageal sphincter (LES). Vasoactive intestinal peptide (VIP) caused dose-dependent relaxation of LES, and H2O2 reduced VIP-induced relaxation. Relaxation was also attenuated by pertussis toxin (PTX), indicating a Gi/o component. VIP treatment increased [35S]GTPgammaS binding to Gs and Gi3 protein, but not to Go, Gq, Gil or Gi2. This increase in Gs or Gi3 binding was reduced by H2O2. However, the relaxation induced by sodium nitroprusside (SNP), 3-morpholino sydnomine (SIN-1), 8-br cGMP (cGMP analog), forskolin (adenylate cyclase activator), and dibutyryl-cAMP (a stable cAMP analog) was not reduced by H2O2. These data suggest that H202 inhibits VIP-induced relaxation via a Gi-dependent pathway, perhaps by inhibiting the activation of G(i3) or Gs downstream of the VIP receptor and independent of cAMP or NO-cGMP signaling.


Asunto(s)
Esfínter Esofágico Inferior/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Relajación Muscular/efectos de los fármacos , Péptido Intestinal Vasoactivo/farmacología , Animales , Gatos , Colforsina/farmacología , GMP Cíclico/fisiología , Relación Dosis-Respuesta a Droga , Esfínter Esofágico Inferior/fisiología , Femenino , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino
10.
J Drug Target ; 25(6): 523-531, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28140690

RESUMEN

Notch pathway was found to be activated in most glioblastomas (GBMs), underlining the importance of Notch in formation and recurrence of GBM. In this study, a Notch inhibitory peptide, dominant negative MAML (dnMAML), was conjugated to elastin-like polypeptide (ELP) for tumor targeted delivery. ELP is a thermally responsive polypeptide that can be actively and passively targeted to the tumor site by localized application of hyperthermia. This complex was further modified with the addition of a cell penetrating peptide, SynB1, for improved cellular uptake and blood-brain barrier penetration. The SynB1-ELP1-dnMAML was examined for its cellular uptake, cytotoxicity, apoptosis, cell cycle inhibition and the inhibition of target genes' expression. SynB1-ELP1-dnMAML inhibited the growth of D54 and U251 cells by inducing apoptosis and cell cycle arrest, especially in the presence of hyperthermia. Hyperthermia increased overall uptake of the polypeptide by the cells and enhanced the resulting pharmacological effects of dnMAML, showing the inhibition of targets of Notch pathway such as Hes-1 and Hey-L. These results confirm that dnMAML is an effective Notch inhibitor and combination with ELP may allow thermal targeting of the SynB1-ELP1-dnMAML complex in cancer cells while avoiding the dangers of systemic Notch inhibition.


Asunto(s)
Péptidos de Penetración Celular/administración & dosificación , Proteínas de Unión al ADN/administración & dosificación , Glioblastoma/tratamiento farmacológico , Receptores Notch/antagonistas & inhibidores , Factores de Transcripción/administración & dosificación , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Péptidos de Penetración Celular/farmacología , Proteínas de Unión al ADN/farmacocinética , Proteínas de Unión al ADN/farmacología , Sistemas de Liberación de Medicamentos , Elastina/administración & dosificación , Glioblastoma/patología , Humanos , Hipertermia Inducida/métodos , Péptidos/administración & dosificación , Factores de Transcripción/farmacocinética , Factores de Transcripción/farmacología
11.
Cancer Chemother Pharmacol ; 57(6): 811-8, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16175393

RESUMEN

Hybrid biosynthetic approach produced a new anthracycline ID6105 (11-hydroxyaclacinomycin X, Hyrubicin), which has potent antitumor activities against a broad range of cancer cell lines. Like other anthracyclines, ID6105 has the inhibitory effects on DNA synthesis as well as topoisomerase II. As preclinical studies of ID6105, we investigated ID6105's efficacy on human tumors, and cardiotoxicity. In human tumor xenografts, the ID6105's antitumor effects were greater than other anticancer drugs. ID6105 induced tumor regression in Hep G2 human hepatoma model, and slowed down the tumor growth rates in several tumor models. Doxorubicin-refractory tumors such as PC-3, DU-145, and CX-1 were sensitive to ID6105, and the growth of EKVX, lung cancer, which did not respond to paclitaxel, was also inhibited by ID6105, but tumor mass in CFPA, MCF7, and HCT-15 was not reduced by ID6105. The cardiotoxicity of ID6105 has also been assessed in rats. ID6105 did not induce any remarkable histopathological changes in hearts, and its lipid peroxidation in rat cardiac muscles did not occur as much as doxorubicin, indicating that the cardiotoxicity of ID6105 is remarkably lower than that of doxorubicin. Taking all into account, our results suggest that ID6105 would be a promising candidate for a novel anthracycline chemotherapeutic agent.


Asunto(s)
Aclarubicina/análogos & derivados , Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Corazón/efectos de los fármacos , Inhibidores de la Síntesis del Ácido Nucleico/uso terapéutico , Aclarubicina/uso terapéutico , Animales , Línea Celular Tumoral , Femenino , Humanos , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos BALB C , Miocardio/metabolismo , Miocardio/patología , Miocardio/ultraestructura , Ratas , Ratas Sprague-Dawley , Inhibidores de Topoisomerasa II , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
World J Gastroenterol ; 12(14): 2259-63, 2006 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-16610033

RESUMEN

AIM: To investigate the mechanism of bombesin-induced circular smooth muscle cell contraction in cat esophagus. METHODS: Specific G protein or phospholipase C involved in cat esophagus contraction was identified, muscle cells were permeabilized with saponin. After permeabilization of muscle cells, the Gi3 antibody inhibited bombesin-induced smooth muscle cell contraction. RESULTS: Incubation of permeabilized circular muscle cells with PLC-beta3 antibody could inhibit bombesin-induced contraction. H-7, chelerythrine (PKC inhibitor) and genistein (protein tyrosine kinase inhibitor) inhibited bombesin-induced contraction, but DAG kinase inhibitor, R59949, could not inhibit it. To examine which mitogen-activated protein kinase (MAPK) was involved in bombesin-induced contraction, the specific MAPK inhibitors (MEK inhibitor, PD98059 and p38 MAPK inhibitor, SB202190) were used. Preincubation of PD98059 blocked the contraction induced by bombesin in a concentration-dependent manner. However, SB202190 had no effects on contraction. CONCLUSION: Bombesin-induced circular muscle cell contraction in cat esophagus is madiated via a PKC or a PTK-dependent pathway or p44/p42 MAPK pathway.


Asunto(s)
Bombesina/farmacología , Esófago/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Transducción de Señal/fisiología , Animales , Gatos , Esófago/fisiología , Isoenzimas/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Miocitos del Músculo Liso/fisiología , Fosfolipasa C beta , Proteína Quinasa C/fisiología , Receptores Acoplados a Proteínas G/fisiología , Fosfolipasas de Tipo C/fisiología
13.
Arch Pharm Res ; 28(4): 476-82, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15918523

RESUMEN

We investigated the pharmacokinetics of 11-hydroxyaclacinomycin X (ID-6105), a novel anthracycline, after intravenous (i.v.) bolus administration at a multiple dose every 24 h for 5 days in rats. To analyze ID-6105 levels in biological samples, we used an HPLC-based method which was validated in a pharmacokinetic study by suitable criteria. The concentrations of ID-6105 after the multiple administration for 5 days were not significantly different from the results after the single administration. The t1/2alpha, t1/2beta, Vdss, and CLt after the multiple administration were not significantly different from the values after the single administration. Moreover, the concentrations of ID-6105 1 min at day 1-5 after i.v. bolus multiple administration did not show the significant difference. Of the various tissues, ID-6105 mainly distributed to the kidney, lung, spleen, adrenal gland, and liver after i.v. bolus multiple administration. ID-6105 concentrations in the kidney or lung 2 h after i.v. bolus administration were comparable to the plasma concentration shortly after i.v. bolus administration. However, the ID-6105 concentrations in various tissues 48 h after i.v. bolus administration decreased to low levels. ID-6105 was excreted largely in the bile after i.v. bolus multiple administration at the dose of 3 mg/kg. The amounts of ID-6105 found in the bile by 12 h or in the urine by 48 h after the administration were calculated to be 14.1% or 4.55% of the initial dose, respectively, indicating that ID-6105 is mostly excreted in the bile. In conclusion, ID-6105 was rapidly cleared from the blood and transferred to tissues, suggesting that ID-6105 might not be accumulated in the blood following i.v. bolus multiple dosages of 3 mg/kg every 24 h for 5 days. By 48 h after i.v. bolus administration, ID-6105 concentrations in various tissues had decreased to very low levels. The majority of ID-6105 appears to be excreted in the bile.


Asunto(s)
Aclarubicina/análogos & derivados , Aclarubicina/farmacocinética , Antibióticos Antineoplásicos/farmacocinética , Aclarubicina/administración & dosificación , Aclarubicina/sangre , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/sangre , Disponibilidad Biológica , Cromatografía Líquida de Alta Presión , Esquema de Medicación , Inyecciones Intravenosas , Masculino , Tasa de Depuración Metabólica , Ratas , Ratas Sprague-Dawley , Distribución Tisular
14.
Trends Mol Med ; 21(9): 560-70, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26186888

RESUMEN

Cell-penetrating peptides (CPP) provide an efficient strategy for the intracellular delivery of bioactive molecules in various biomedical applications. This review focuses on recent advances in the use of CPPs to deliver anticancer therapeutics and imaging reagents to cancer cells, along with CPP contributions to novel tumor-targeting techniques. CPPs are now used extensively to deliver a variety of therapeutics, despite lacking cell specificity and having a short duration of action. Resolution of these shortcomings to enable increased cancer cell and/or tumor specificity could improve CPP-based drug delivery strategies, expand combined drug delivery possibilities, and strengthen future clinical applications of these peptides.


Asunto(s)
Antineoplásicos/administración & dosificación , Péptidos de Penetración Celular/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Animales , Humanos
15.
Expert Opin Drug Deliv ; 12(4): 653-67, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25350837

RESUMEN

INTRODUCTION: Despite their poor specificity, small molecule drugs are considered more powerful and effective than other current chemotherapies. A promising method for targeting these anticancer drugs to tumors, elastin-like polypeptides (ELP), has recently emerged. When an anticancer drug that has been conjugated to an ELP is administered, and focal hyperthermia applied, the thermoresponsive properties and enhanced permeability and retention effects of the ELP facilitate drug aggregation within tumor tissues. By incorporating a cell penetrating peptide onto this ELP-chemotherapeutic construct, even greater drug uptake into tumor cells can be achieved. AREAS COVERED: The review explores the preclinical study progress of ELP-based drug delivery technology and discusses its potential in cancer therapy. Recent experimental work has shown that a delivery construct consisting of an ELP-therapeutic peptide (e.g., the c-Myc-inhibitory peptide, or the p21(WAF1/CIP1)-derived peptide), as well as ELP-small molecule drugs (e.g., doxorubicin, paclitaxel), can be thermally targeted to accumulate in tumors and diminish their growth. EXPERT OPINION: ELP drug delivery technology is complementary and synergistic to current drug delivery modalities and based on existing hyperthermia technology. By using this technology to achieve chemotherapeutic targeting, efficacy can be improved and side effects reduced in comparison with current regimens, providing treatment alternatives and/or augmenting current therapies for cancer treatment.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Doxorrubicina/administración & dosificación , Doxorrubicina/uso terapéutico , Elastina/química , Calor , Humanos , Neoplasias/patología , Paclitaxel/administración & dosificación , Paclitaxel/uso terapéutico , Péptidos/química , Permeabilidad
16.
Eur J Pharm Biopharm ; 88(2): 382-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24866938

RESUMEN

Elastin-like polypeptides (ELP) are thermally responsive polypeptides that are soluble in solutions at 37°C, but which aggregate above 42°C. ELP can be used as effective carrier systems of anticancer molecules, because they can be targeted to tumor sites through the application of local hyperthermia. Since molecular size largely influences how successfully therapeutic agents can cross the vasculatures of tumors, it was crucial to determine an optimal molecular size. In this study, we designed and evaluated three ELP macromolecules with varying molecular weights (43, 63, and 122 kDa), with the goal of determining which would optimize the ELP drug delivery system. The N-terminus of the ELP macromolecule was modified with the cell penetrating peptide Bac to enhance intratumoral and intracellular uptake, and it was also confirmed that each polypeptide had the target transition temperature of 37-42°C and the results of the studies, using tumor-bearing mice, showed that the tumor accumulations increased in the case of all three peptides when local hyperthermia was applied, but that the elimination patterns from these tumors varied according to peptide size. Local hyperthermia was found to produce prolonged retention of all ELP conjugates in tumors except Bac-ELP43. In addition, the pharmacokinetic analysis showed that two larger polypeptides with 63 and 122 kDa have increased AUC in comparison with the 43 kDa polypeptide. These results suggest that, when combined with local hyperthermia, the larger ELP conjugates (63 and 122 kDa) have advantages over the smaller Bac-ELP43 polypeptide in terms of enhanced permeability and higher retention effects.


Asunto(s)
Elastina/metabolismo , Hipertermia Inducida , Neoplasias Experimentales/metabolismo , Péptidos/metabolismo , Animales , Elastina/química , Elastina/farmacocinética , Ratones , Microscopía Fluorescente , Peso Molecular , Neoplasias Experimentales/patología , Péptidos/química , Péptidos/farmacocinética
17.
Cancer Lett ; 348(1-2): 177-84, 2014 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-24680816

RESUMEN

This work describes the effects of elastin-like polypeptide (ELP) with the p21(Waf1/Cip1)-derived cell cycle inhibitory peptide (p21) on pancreatic tumor cells with gemcitabine. The thermo-responsive property of ELP permits use of a mild, local hyperthermia to target tumors for the transport of chemotherapeutics. In this study, a p21-ELP construct with Bac cell penetrating peptide was designed, and its anticancer activities in pancreatic cancer cell lines was examined. In combination with gemcitabine, the peptide demonstrated enhanced in vitro cytotoxicity as well as tumor growth inhibition in an animal model. Our data suggest that this ELP construct, with gemcitabine, may improve pancreatic cancer therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Hipotermia Inducida , Neoplasias Pancreáticas/tratamiento farmacológico , Transporte Activo de Núcleo Celular , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Péptidos de Penetración Celular/metabolismo , Péptidos de Penetración Celular/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/farmacología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Elastina/metabolismo , Elastina/farmacología , Estudios de Factibilidad , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Fosforilación , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Proteína de Retinoblastoma/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
18.
Ther Deliv ; 5(4): 429-45, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24856169

RESUMEN

Therapeutic peptides offer important cancer treatment approaches. Designed to inhibit oncogenes and other oncoproteins, early therapeutic peptides applications were hampered by pharmacokinetic properties now addressed through tumor targeting strategies. Active targeting with environmentally responsive biopolymers or macromolecules enhances therapeutics accumulation at tumor sites; passive targeting with macromolecules, or liposomes, exploits angiogenesis and poor lymphatic drainage to preferentially accumulate therapeutics within tumors. Genetically engineered, thermally-responsive, elastin-like polypeptides use both strategies and cell-penetrating peptides to further intratumoral cell uptake. This review describes the development and application of cell-penetrating peptide-elastin-like polypeptide therapeutics for the thermally targeted delivery of therapeutic peptides.


Asunto(s)
Antineoplásicos/metabolismo , Permeabilidad de la Membrana Celular , Membrana Celular/metabolismo , Péptidos de Penetración Celular/metabolismo , Portadores de Fármacos , Elastina/metabolismo , Neoplasias/metabolismo , Temperatura , Animales , Antineoplásicos/química , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/genética , Química Farmacéutica , Elastina/química , Elastina/genética , Humanos , Ingeniería de Proteínas , Tecnología Farmacéutica/métodos
19.
Drug Des Devel Ther ; 8: 1649-58, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25336913

RESUMEN

Current therapies for the treatment of pancreatic cancer are limited. The limitations of this type of treatment are abundant. The majority of chemotherapeutic agents used in clinics are highly toxic to both tumor cells and normal tissues due to the lack of specificity. Resistance can develop due to overexposure of these agents. To address these issues, these agents must be made more exclusive toward the tumor site. We have developed a macromolecular carrier based on the sequence of the biopolymer elastin-like polypeptide (ELP) that is able to aggregate upon reaching the externally heated tumor environment. This carrier is specific to the tumor as it only aggregates at the heated tumor site. ELP is soluble below its transition temperature but will aggregate when the temperature is raised above its transition temperature. ELP was modified by p21, a cell cycle inhibitory peptide, and the addition of Bac, a cell-penetrating peptide with nuclear localization capabilities. In this study, p21-ELP-Bac and its control, ELP-p21, were used in cell proliferation studies using the pancreatic cancer cell lines Panc-1, MiaPaca-2, and S2013. ELP-p21 had little effect on proliferation, while the half maximal inhibitory concentration of p21-ELP-Bac was ∼30 µM. As translocation across the plasma membrane is a limiting step for delivery of macromolecules, these polypeptides were utilized in a pancreatic xenograft model to study the plasma clearance, biodistribution, tumor accumulation, and tumor reduction capabilities of the polypeptide with and without a cell-penetrating peptide.


Asunto(s)
Biopolímeros/química , Péptidos de Penetración Celular/química , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Neoplasias Experimentales/metabolismo , Neoplasias Pancreáticas/metabolismo , Temperatura , Animales , Biopolímeros/administración & dosificación , Proliferación Celular/efectos de los fármacos , Péptidos de Penetración Celular/administración & dosificación , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Relación Estructura-Actividad , Células Tumorales Cultivadas
20.
Anticancer Res ; 30(11): 4607-12, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21115913

RESUMEN

Indole-3-acetic acid (IAA) has recently shown anticancer activity in combination with horseradish peroxidase. The current study demonstrated that IAA irradiated with ultraviolet B (IAA(UVB)) is able to generate free radicals and induce cell death in a time-dependent fashion in PC-3 prostate cancer cells, while PC-3 cells treated with IAA alone exhibited no toxic responses. It was also found through Western blot analysis that the cytotoxic effect of IAA(UVB) resulted from apoptosis. Treatment with IAA(UVB) for 24 hours showed a significant increase in phosphorylated p38 mitogen-activated protein kinase and c-Jun N-terminal kinase, the stress signaling proteins. Furthermore, pro-caspases (-3, -8, and -9) were clearly down-regulated and poly(ADP-ribose) polymerase cleavages were demonstrated in the group treated with IAA(UVB). Flow cytometric analysis also demonstrated the induction of apoptosis by IAA(UVB) in PC-3 cells. In conclusion, this study demonstrated that IAA induced cell death in combination with UVB irradiation by increasing apoptosis in PC-3 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Ácidos Indolacéticos/farmacología , Neoplasias de la Próstata/patología , Rayos Ultravioleta , Western Blotting , Caspasas/metabolismo , Línea Celular Tumoral , Terapia Combinada , Citometría de Flujo , Radicales Libres/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Poli(ADP-Ribosa) Polimerasas/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/radioterapia , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA