Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
FASEB J ; 36(1): e22090, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34907595

RESUMEN

Despite many advances in infection control practices, including prophylactic antibiotics, surgical site infections (SSIs) remain a significant cause of morbidity, prolonged hospitalization, and death worldwide. Our innate immune system possesses a multitude of powerful antimicrobial strategies which make it highly effective in combating bacterial, fungal, and viral infections. However, pathogens use various stealth mechanisms to avoid the innate immune system, which in turn buy them time to colonize wounds and damage tissues at surgical sites. We hypothesized that immunomodulators that can jumpstart and activate innate immune responses at surgical sites, would likely reduce infection at surgical sites. We used three immunomodulators; fMLP (formyl-Methionine-Lysine-Proline), CCL3 (MIP-1α), and LPS (Lipopolysaccharide), based on their documented ability to elicit strong inflammatory responses; in a surgical wound infection model with Pseudomonas aeruginosa to evaluate our hypothesis. Our data indicate that one-time topical treatment with these immunomodulators at low doses significantly increased proinflammatory responses in infected and uninfected surgical wounds and were as effective, (or even better), than a potent prophylactic antibiotic (Tobramycin) in reducing P. aeruginosa infection in wounds. Our data further show that immunomodulators did not have adverse effects on tissue repair and wound healing processes. Rather, they enhanced healing in both infected and uninfected wounds. Collectively, our data demonstrate that harnessing the power of the innate immune system by immunomodulators can significantly boost infection control and potentially stimulate healing. We propose that topical treatment with these immunomodulators at the time of surgery may have therapeutic potential in combating SSI, alone or in combination with prophylactic antibiotics.


Asunto(s)
Factores Inmunológicos/farmacología , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/inmunología , Infección de la Herida Quirúrgica/tratamiento farmacológico , Animales , Evaluación de Medicamentos , Ratones , Ratones Noqueados , Infecciones por Pseudomonas/inmunología , Infección de la Herida Quirúrgica/inmunología , Infección de la Herida Quirúrgica/microbiología
2.
J Chem Inf Model ; 61(11): 5469-5483, 2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34666487

RESUMEN

COVID-19, an acute viral pneumonia, has emerged as a devastating pandemic. Drug repurposing allows researchers to find different indications of FDA-approved or investigational drugs. In this current study, a sequence of pharmacophore and molecular modeling-based screening against COVID-19 Mpro (PDB: 6LU7) suggested a subset of drugs, from the Drug Bank database, which may have antiviral activity. A total of 44 out of 8823 of the most promising virtual hits from the Drug Bank were subjected to molecular dynamics simulation experiments to explore the strength of their interactions with the SARS-CoV-2 Mpro active site. MD findings point toward three drugs (DB04020, DB12411, and DB11779) with very low relative free energies for SARS-CoV-2 Mpro with interactions at His41 and Met49. MD simulations identified an additional interaction with Glu166, which enhanced the binding affinity significantly. Therefore, Glu166 could be an interesting target for structure-based drug design. Quantitative structural-activity relationship analysis was performed on the 44 most promising hits from molecular docking-based virtual screening. Partial least square regression accurately predicted the values of independent drug candidates' binding energy with impressively high accuracy. Finally, the EC50 and CC50 of 10 drug candidates were measured against SARS-CoV-2 in cell culture. Nilotinib and bemcentinib had EC50 values of 2.6 and 1.1 µM, respectively. In summary, the results of our computer-aided drug design provide a roadmap for rational drug design of Mpro inhibitors and the discovery of certified medications as COVID-19 antiviral therapeutics.


Asunto(s)
COVID-19 , Inhibidores de Proteasas , Antivirales/farmacología , Proteasas 3C de Coronavirus , Reposicionamiento de Medicamentos , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Pirimidinas , SARS-CoV-2
3.
Int J Cancer ; 141(9): 1912-1920, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28635011

RESUMEN

Chemotherapy have commonly been used in maximum tolerated dose to completely eradicate the cancer. However, such treatments often failed due to the complex and dynamic nature of cancer. Therefore, it has been suggested that cancer should be treated as a chronic disease, controlling its growth by providing continuous therapeutic pressure for long-term. Such an approach, however, requires a therapy that is non-toxic and orally available with sufficient potency. Herein, we propose a radiotherapy-assisted orally available metronomic apoptosis-targeted chemotherapy, which delivers doxorubicin continuously to the irradiated tumor with high selectivity while causing minimal toxicities to the normal tissues. DEVD-S-DOX/DCK complex is the anticancer prodrug for our strategy that could selectively release doxorubicin in the irradiated tumor tissue with sufficient oral bioavailability. The prodrug was completely inactive by itself, but displayed potent anticancer activity when coupled with radiotherapy. Consequently, the daily oral administration of DEVD-S-DOX/DCK in combination with the low-dose radiotherapy effectively suppressed the growth of tumor in vivo with no significant systemic toxicities despite that the accumulated dose of doxorubicin exceeded 150 mg/kg. Therefore, the our novel therapy using DEVD-S-DOX/DCK complex is considered as an outstanding treatment option for treating cancer for long-term attributed to its oral availability and low-toxicity profile as well as the potent anticancer effect.


Asunto(s)
Doxorrubicina/administración & dosificación , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Profármacos/administración & dosificación , Administración Oral , Animales , Apoptosis/efectos de los fármacos , Células CACO-2 , Terapia Combinada , Doxorrubicina/química , Sistemas de Liberación de Medicamentos , Humanos , Dosis Máxima Tolerada , Ratones , Neoplasias/patología , Profármacos/química , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Pharm ; 12(11): 3935-42, 2015 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-26448404

RESUMEN

Low molecular weight heparin (LMWH) and its derivatives have been reported to possess antiangiogenic effect via electrostatic interaction with various angiogenic growth factors such as VEGF165. However, clinical applications of LMWH for anticancer therapy have been restricted due to its anticoagulant effect and insufficient therapeutic efficacy. To overcome these limitations and enhance the antiangiogenic efficacy, LMWH was conjugated with suramin fragments that have a binding affinity to the heparin-binding domain (HBD) of proteins. The conjugation of suramin fragments to LMWH enhanced the antiangiogenic effect of LMWH by increasing the binding affinity to VEGF165, while decreasing its anticoagulant activity. The chemical conjugate of LMWH and suramin fragments (LHsura) showed a substantial inhibitory effect on VEGF165-mediated cell proliferation, migration, and tube formation of HUVECs without significant cytotoxicity in vitro. Finally, we confirmed the anticancer effect of LHsura (61.4% vs control) in a SCC7-bearing mouse model.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Heparina de Bajo-Peso-Molecular/química , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Suramina/química , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Carcinoma de Células Escamosas/patología , Proliferación Celular/efectos de los fármacos , Simulación por Computador , Heparina de Bajo-Peso-Molecular/administración & dosificación , Humanos , Ratones , Ratones Endogámicos C3H , Suramina/administración & dosificación , Resonancia por Plasmón de Superficie , Células Tumorales Cultivadas , Cicatrización de Heridas/efectos de los fármacos
5.
Mol Pharm ; 12(6): 1911-20, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25892399

RESUMEN

Currently, oral administration of insulin still remains the best option to avoid the burden of repeated subcutaneous injections and to improve its pharmacokinetics. The objective of the present investigation was to demonstrate the absorption mechanism of insulin in the physical complexation of deoxycholyl-l-lysyl-methylester (DCK) for oral delivery. The oral insulin/DCK complex was prepared by making a physical complex of insulin aspart with DCK through ion-pair interaction in water. For the cellular uptake study, fluorescein-labeled insulin or DCK were prepared according to a standard protocol and applied to Caco-2 or MDCK cell lines. For the PK/PD studies, we performed intrajejunal administration of different formulation of insulin/DCK complex to diabetic rats. The resulting insulin and DCK complex demonstrated greatly enhanced lipophilicity as well as increased permeation across Caco-2 monolayers. The immunofluorescence study revealed the distribution of the complex in the cytoplasm of Caco-2 cells. Moreover, in the apical sodium bile acid transporter (ASBT) transfected MDCK, the insulin/DCK complex showed interaction with ASBT, and also demonstrated absorption through passive diffusion. We could not find that any evidence of endocytosis in relation to the uptake of insulin complex in vitro. In the rat intestine model, the highest absorption of insulin complex was observed in the jejunum at 1 h and then in the ileum at 2-4 h. In PK/PD study, the complex showed a similar PK profile to that of SC insulin. Overall, the study showed that the effect of DCK on enhancing the absorption of insulin resulted from transcellular processes as well as bile acid transporter activity.


Asunto(s)
Ácido Quenodesoxicólico/análogos & derivados , Portadores de Fármacos/química , Insulina/química , Intestino Delgado/metabolismo , Lisina/análogos & derivados , Administración Oral , Animales , Células CACO-2 , Ácido Quenodesoxicólico/química , Ácido Quenodesoxicólico/farmacocinética , Perros , Portadores de Fármacos/farmacocinética , Humanos , Insulina/farmacocinética , Yeyuno/metabolismo , Lisina/química , Lisina/farmacocinética , Células de Riñón Canino Madin Darby , Masculino , Ratas , Ratas Sprague-Dawley
6.
J Invest Dermatol ; 144(2): 387-397.e11, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37619833

RESUMEN

Diabetic chronic ulcers are plagued with persistent nonresolving inflammation. However, diabetic wound environment early after injury suffers from inadequate inflammatory responses due to reductions in proinflammatory cytokines levels. Diabetic neutrophils have known impairments in bactericidal functions. We hypothesized that reduced bacterial killing by diabetic neutrophils, due to their bactericidal functional impairments, results in reduced bioactive bacterial products, known as pathogen-associated molecular patterns, which in turn contribute to reduced signaling through toll-like receptors, leading to inadequate production of proinflammatory cytokines in infected diabetic wound early after injury. We tested our hypothesis in db/db type 2 obese diabetic mouse wound infection model with Pseudomonas aeruginosa. Our data indicate that despite substantially higher levels of infection, toll-like receptor 4-mediated signaling is reduced in diabetic wounds early after injury owing to reduced bioactive levels of lipopolysaccharide. We further demonstrate that topical treatment with lipopolysaccharide enhances toll-like receptor 4 signaling, increases proinflammatory cytokine production, restores leukocyte trafficking, reduces infection burden, and stimulates healing in diabetic wounds. We posit that lipopolysaccharide may be a viable therapeutic option for the treatment of diabetic foot ulcers if it is applied topically after the surgical debridement process, which is intended to reset chronic ulcers into acute fresh wounds.


Asunto(s)
Diabetes Mellitus Tipo 2 , Pie Diabético , Infección de Heridas , Ratones , Animales , Receptor Toll-Like 4 , Moléculas de Patrón Molecular Asociado a Patógenos/uso terapéutico , Lipopolisacáridos , Infección de Heridas/tratamiento farmacológico , Pie Diabético/tratamiento farmacológico , Antibacterianos/uso terapéutico , Inmunidad , Citocinas
7.
Mol Cancer Ther ; 21(7): 1103-1114, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35499388

RESUMEN

Triple-negative breast cancer (TNBC) is a highly aggressive type of breast cancer. Unlike other subtypes of breast cancer, TNBC lacks hormone and growth factor receptor targets. Colchicine-binding site inhibitors (CBSI) targeting tubulin have been recognized as attractive agents for cancer therapy, but there are no CBSI drugs currently FDA approved. CH-2-77 has been reported to have potent antiproliferative activity against a panel of cancer cells in vitro and efficacious antitumor effects on melanoma xenografts, yet, its anticancer activity specifically against TNBC is unknown. Herein, we demonstrate that CH-2-77 inhibits the proliferation of both paclitaxel-sensitive and paclitaxel-resistant TNBC cells with an average IC50 of 3 nmol/L. CH-2-77 also efficiently disrupts the microtubule assembly, inhibits the migration and invasion of TNBC cells, and induces G2-M cell-cycle arrest. The increased number of apoptotic cells and the pattern of expression of apoptosis-related proteins in treated MDA-MB-231 cells suggest that CH-2-77 induces cell apoptosis through the intrinsic apoptotic pathway. In vivo, CH-2-77 shows acceptable overall pharmacokinetics and strongly suppresses the growth of orthotopic MDA-MB-231 xenografts without gross cumulative toxicities when administered 5 times a week. The in vivo efficacy of CH-2-77 (20 mg/kg) is comparable with that of CA4P (28 mg/kg), a CBSI that went through clinical trials. Importantly, CH-2-77 prevents lung metastasis originating from the mammary fat pad in a dose-dependent manner. Our data demonstrate that CH-2-77 is a promising new generation of tubulin inhibitors that inhibit the growth and metastasis of TNBC, and it is worthy of further development as an anticancer agent.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Apoptosis , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular , Colchicina/farmacología , Colchicina/uso terapéutico , Humanos , Paclitaxel/farmacología , Neoplasias de la Mama Triple Negativas/patología , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/farmacología , Moduladores de Tubulina/uso terapéutico
8.
J Med Chem ; 64(17): 13072-13095, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34406768

RESUMEN

Small molecules that interact with the colchicine binding site in tubulin have demonstrated therapeutic efficacy in treating cancers. We report the design, syntheses, and antitumor efficacies of new analogues of pyridopyrimidine and hydroquinoxalinone compounds with improved drug-like characteristics. Eight analogues, 5j, 5k, 5l, 5m, 5n, 5r, 5t, and 5u, showed significant improvement in metabolic stability and demonstrated strong antiproliferative potency in a panel of human cancer cell lines, including melanoma, lung cancer, and breast cancer. We report crystal structures of tubulin in complex with five representative compounds, 5j, 5k, 5l, 5m, and 5t, providing direct confirmation for their binding to the colchicine site in tubulin. A quantitative structure-activity relationship analysis of the synthesized analogues showed strong ability to predict potency. In vivo, 5m (4 mg/kg) and 5t (5 mg/kg) significantly inhibited tumor growth as well as melanoma spontaneous metastasis into the lung and liver against a highly paclitaxel-resistant A375/TxR xenograft model.


Asunto(s)
Antineoplásicos/farmacología , Quinoxalinas/farmacología , Moduladores de Tubulina/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Diseño de Fármacos , Resistencia a Antineoplásicos , Femenino , Humanos , Masculino , Ratones , Relación Estructura-Actividad Cuantitativa , Quinoxalinas/química , Moduladores de Tubulina/química , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Lett ; 495: 76-88, 2020 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-32920198

RESUMEN

Lung cancer is the most common cause of cancer associated mortality. Chemotherapeutic agents, such as paclitaxel, are important treatment options but drug resistance often develops upon prolonged use. We report here the preclinical evaluation of a new orally available tubulin inhibitor, VERU-111, which can overcome several ABC-transporters mediated multi-drug resistance associated with taxane treatment. In vitro, VERU-111 prevents cell proliferation, invasion, migration and colony formation in both paclitaxel-sensitive and paclitaxel-resistant A549 lung cancer cells. VERU-111 effectively inhibits tubulin polymerization, arrests cells in G2/M phase, and induces cancer cell apoptosis. Further evaluation of various apoptotic proteins revealed that treatment of VERU-111 increases the expression of cleaved-PARP, cleaved-caspase-3 and p-histone H3 proteins. In vivo, orally administered VERU-111 in a paclitaxel-sensitive A549 xenograft model strongly inhibits tumor growth in a dose-dependent manner and is equally potent with paclitaxel. When tested in a highly paclitaxel-resistant A549/TxR tumor model, VERU-111 is as effective as the parental A549 model in significantly reducing the tumor volume, whereas paclitaxel is essentially ineffective. Collectively, this study showed that VERU-111 is a promising new generation of anti-tubulin agent for the treatment of taxane-resistant lung cancer.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Imidazoles/administración & dosificación , Indoles/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Paclitaxel/administración & dosificación , Moduladores de Tubulina/administración & dosificación , Células A549 , Administración Oral , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos , Imidazoles/farmacología , Indoles/farmacología , Neoplasias Pulmonares/metabolismo , Ratones , Paclitaxel/farmacología , Moduladores de Tubulina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Biomaterials ; 182: 35-43, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30103170

RESUMEN

Metronomic chemotherapy, which is defined as a low-dose and frequent administration of cytotoxic drugs without drug-free breaks, has been recently emerged as an alternative to traditional MTD therapy and has shown therapeutic benefit in breast cancer patients in numbers of clinical studies. Unlike MTD, metronomic chemotherapy acts by multiple mechanisms including antiangiogenic effect and immunomodulation, but the direct cytotoxic effect only playing a minor role due to the lowered dose. In this light, within the limits of p53-deficient breast cancer, we demonstrate the enhanced anticancer effect of metronomic chemotherapy using doxorubicin when combined with Chk1 inhibitor MK-8776 by specifically augmenting the direct cytotoxic effect on cancer cells. Since the oral drug is greatly favored in metronomic chemotherapy due to the frequent and potential long-term administration, we prepared an oral doxorubicin by producing an ionic complex with deoxycholic acid, which showed sufficient bioavailability and anticancer effect when administered orally. MK-8776 selectively enhanced the cytotoxic effect of low-concentration doxorubicin in p53-deficient breast cancer cells by abrogating the Chk1-dependent cell cycle arrest in vitro. Consistently, combining MK-8776 significantly improved the anticancer effect of the daily administered oral doxorubicin in p53-deficient breast cancer xenografts especially in a lower dose of doxorubicin without evident systemic toxicities. Combination therapy of MK-8776 and metronomic oral doxorubicin would be thus promising in the treatment of p53-deficient breast cancer benefited from the augmented direct cytotoxic effect and low risk of toxicities.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Doxorrubicina/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Proteína p53 Supresora de Tumor/genética , Administración Metronómica , Administración Oral , Animales , Antibióticos Antineoplásicos/uso terapéutico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Doxorrubicina/uso terapéutico , Femenino , Eliminación de Gen , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico
11.
J Control Release ; 284: 160-170, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-29908222

RESUMEN

There is a growing interest in preclinical research to consider low-dose metronomic chemotherapy as antiangiogenic cancer treatment. Oral metronomic therapy, in particular, has shown much promise with its ease of daily administration and higher therapeutics window. In that regard, we developed oral pemetrexed using the physical complex with the bile acid enhancers (DCK). In a caco-2 permeability study, the oral pemetrexed/DCK complex had significantly higher drug uptake, and inhibited efflux transporter activity as well. We further observed that the mechanism of oral drug uptake was related to transcellular along with bile acid transporter mediated pathways. The oral administration of drug complex in rats revealed high bioavailability (22.37%) and extended mean residence time. Using SCC7 and A549 xenograft models, we demonstrated that antitumor effects from daily oral metronomic pemetrexed significantly reduced tumor in a dose-dependent manner. The antitumor activity of oral pemetrexed/DCK complex plus cisplatin was superior to both monotherapies. The xenograft study also revealed that oral metronomic therapy markedly reduced microvessel density, proliferation and increased apoptosis in the tumor tissues. Oral metronomic doses were significantly correlated with the elevation of plasma deoxyuridine level, an essential biomarker for pemetrexed therapy. One-month toxicity study confirmed that daily dosing of oral pemetrexed is safe by investigating apoptosis in the gut tissues from mice. Moreover, we analyzed different biochemical parameters and enzymes from the blood to prove that our newly developed oral pemetrexed complex is well tolerated.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Pemetrexed/administración & dosificación , Células A549 , Administración Metronómica , Administración Oral , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Apoptosis , Ácidos y Sales Biliares/química , Células CACO-2 , Proliferación Celular/efectos de los fármacos , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Pemetrexed/farmacocinética , Pemetrexed/uso terapéutico , Vehículos Farmacéuticos/química , Ratas Sprague-Dawley
12.
Biomaterials ; 139: 56-66, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28586719

RESUMEN

Clinical studies have found that the incidence of cancer metastasis through the lymphatic vessels are 3-5 times higher than that through the blood vessels. These findings suggest the potency of anti-lymphangiogenic therapy in reducing the incidence of cancer metastasis. Previously, we reported LHbisD4, which is the conjugate of low molecular weight heparin (LMWH) and four bis-deoxycholates as a potent anti-angiogenic drug with less toxicity and orally active property. Here, we show that LHbisD4 could also suppress the formation of new lymphatic vessels and attenuate the incidence of metastasis by blocking VEGF-C signaling pathway. LHbisD4 significantly enhanced binding affinity with VEGF-C when compared with LMWH, which enables LHbisD4 to suppress the proliferation, migration and formation of tubular structures of human dermal lymphatic endothelial cells(HDLECs) in in vitro condition even in the presence of excessive amounts of VEGF-C. Similarly, we found that the density of lymphatic vessels in the primary tumor tissue in breast cancer bearing mice was significantly diminished when LHbisD4 was administered compared with the control group. Also, the incidence of axillary lymph nodes and distant organ metastasis was significantly reduced in the LHbisD4 administered group, which demonstrates that LHbisD4 could successfully lower the incidence of metastasis through blocking VEGF-C induced lymphangiogenesis. Based on these results, we propose LHbisD4 as a potent anti-cancer drug that can reduce the incidence of metastasis by suppressing lymphangiogenesis through blocking VEGF-C signaling pathway.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Heparina de Bajo-Peso-Molecular/química , Linfangiogénesis/efectos de los fármacos , Factor C de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Animales , Movimiento Celular , Proliferación Celular , Ácido Desoxicólico/química , Células Endoteliales/efectos de los fármacos , Femenino , Humanos , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Metástasis Linfática , Vasos Linfáticos/efectos de los fármacos , Vasos Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Neovascularización Patológica/tratamiento farmacológico
13.
J Control Release ; 249: 42-52, 2017 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-28093298

RESUMEN

Metronomic chemotherapy has translated into favorable toxicity profile and capable of delaying tumor progression. Despite its promise, conventional injectable chemotherapeutics are not meaningful to use as metronomic due to the necessity of frequent administration for personalized therapy in long-term cancer treatments. This study aims to exploit the benefits of the oral application of carboplatin as metronomic therapy for non-small cell lung cancer (NSCLC). We developed an orally active carboplatin by physical complexation with a deoxycholic acid (DOCA). The X-ray diffraction (XRD) patterns showed the disappearance of crystalline peaks from carboplatin by forming the complex with DOCA. In vivo pharmacokinetic (PK) study confirmed the oral absorption of carboplatin/DOCA complex. The oral bioavailability of carboplatin/DOCA complex and native carboplatin were calculated as 24.33% and 1.16%, respectively, when a single 50mg/kg oral dose was administered. Further findings of oral bioavailability during a low-dose daily administration of the complex (10mg/kg) for 3weeks were showed 19.17% at day-0, 30.27% at day-7, 26.77% at day-14, and 22.48% at day-21, demonstrating its potential for metronomic chemotherapy. The dose dependent antitumor effects of oral carboplatin were evaluated in SCC7 and A549 tumor xenograft mice. It was found that the oral carboplatin complex exhibited potent anti-tumor activity at 10mg/kg (74.09% vs. control, P<0.01) and 20mg/kg dose (86.22% vs. control, P<0.01) in A549 tumor. The number of TUNEL positive cells in the tumor sections was also significantly increased during oral therapy (3.95% in control, whereas 21.37% and 32.39% in 10mg/kg and 20mg/kg dose, respectively; P<0.001). The enhanced anti-tumor efficacy of oral metronomic therapy was attributed with its antiangiogenic mechanism where new blood vessel formation was notably decreased. Finally, the safety of oral complex was confirmed by three weeks toxicity studies; there were no significant systemic or local abnormalities found in mice at 10mg/kg daily oral dose. Our study thus describes an effective and safe oral formulation of carboplatin as a metronomic chemotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Carboplatino/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Ácido Desoxicólico/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Pulmón/efectos de los fármacos , Administración Metronómica , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica , Carboplatino/administración & dosificación , Carboplatino/análogos & derivados , Carboplatino/farmacocinética , Carcinoma de Pulmón de Células no Pequeñas/patología , Ácido Desoxicólico/administración & dosificación , Ácido Desoxicólico/análogos & derivados , Ácido Desoxicólico/farmacocinética , Pulmón/patología , Neoplasias Pulmonares/patología , Masculino , Ratones Desnudos , Modelos Moleculares , Ratas Sprague-Dawley
14.
Biomaterials ; 86: 56-67, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26890038

RESUMEN

Targeting multiple stages in metastatic breast cancer is one of the effective ways to inhibit metastatic progression. To target human metastatic breast cancer as well as improving patient compliance, we developed an orally active low molecular weight heparin (LMWH)-taurocholate conjugated with tetrameric deoxycholic acid, namely LHTD4, which followed by physical complexation with a synthetic bile acid enhancer, DCK. In breast cancer, both transforming growth factor-ß1 (TGF-ß1) and CXCL12 exhibit enhanced metastatic activity during the initiation and progression stages of breast cancer, thus we direct the focus on investigating the antimetastatic effect of LHTD4/DCK complex by targeting TGF-ß1 and CXCL12. Computer simulation study and SPR analysis were performed for the binding confirmation of LHTD4 with TGF-ß1 and CXCL12. We carried out in vitro phosphorylation assays of the consecutive receptors of TGF-ß1 and CXCL12 (TGF-ß1R1 and CXCR4, respectively). Effects of LHTD4 on in vitro cell migration (induced by TGF-ß1) and chemotaxis (mediated by CXCL12) were investigated. The in vivo anti-metastatic effect of LHTD4 was evaluated in an accelerated metastasis model and an orthotopic MDA-MB-231 breast cancer model. The obtained KD values of TGF-ß1 and CXCL12 with LHTD4 were 0.85 and 0.019 µM respectively. The simulation study showed that binding affinities of LHTD4 fragment with either TGF-ß1 or CXCL12 through additional electrostatic interaction was more stable than that of LMWH fragment. In vitro phosphorylation assays of TGF-ß1R1 and CXCR4 showed that the effective inhibition of receptor phosphorylation was observed with the treatment of LHTD4. The expressions of epithelial to mesenchymal transition (EMT) marker proteins such as vimentin and Snail were prevented by LTHD4 treatment in in vitro studies with TGF-ß1 treated MDA-MB-231 cells. Moreover, we observed that LHTD4 negatively regulated the functions of TGF-ß1 and CXCL12 on migration and invasion of breast cancer cell. In several advanced orthotopic and experimental breast cancer metastasis murine models, the treatment with LHTD4 (5 mg/kg daily, p.o.) significantly inhibited metastasis compared to the control. Overall, LHTD4 exhibited anti-metastatic effects by inhibiting TGF-ß1 and CXCL12, and the clinically relevant dose of orally active LHTD4 was found to be effective in preclinical studies without any apparent toxicity.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Ácido Desoxicólico/uso terapéutico , Heparina de Bajo-Peso-Molecular/uso terapéutico , Metástasis de la Neoplasia/prevención & control , Ácido Taurocólico/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Mama/efectos de los fármacos , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Ácido Desoxicólico/análogos & derivados , Ácido Desoxicólico/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Heparina de Bajo-Peso-Molecular/análogos & derivados , Heparina de Bajo-Peso-Molecular/farmacología , Humanos , Ratones SCID , Terapia Molecular Dirigida , Metástasis de la Neoplasia/patología , Fosforilación/efectos de los fármacos , Ácido Taurocólico/análogos & derivados , Ácido Taurocólico/farmacología , Factor de Crecimiento Transformador beta1/metabolismo
15.
J Control Release ; 197: 180-9, 2015 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-25445701

RESUMEN

Angiogenesis is a key feature of cancer development, thus it is a good target for cancer therapy. However, drugs that have been designed to block angiogenesis mainly capture growth factors in circulation, resulting not only in the transient inhibition of tumor progression but also in producing undesirable side effects. Nanoparticular drug delivery systems, on the other hand, may help overcome such drawbacks and improve the efficacy of anti-angiogenic therapies by altering the biodistribution and pharmacokinetics, improving tumor targeting ability, and reducing side effects. In this light, we propose a new approach of anti-angiogenic therapy that combines strategies of long circulating, passive tumor targeting, and anti-angiogenesis efficacy using a new polyelectrolyte complex system that combines LHT7, a previously developed heparin-based angiogenesis inhibitor, with a protamine to form a self-assembling nanocomplex with a mean diameter of 200nm, which is effective for anti-angiogenesis therapy. At first, LHT7 was modified with polyethylene glycol (PEG). We observed that PEG-LHT7/protamine nanocomplex was stable in buffer and slowly dissociated in plasma (9% dissociation for 24h). Compared to the free form of PEG-LHT7, the mean residence time of PEG-LHT7/protamine nanocomplex was found higher (15.9h) with its increased accumulation in tumor. Most importantly, PEG-LHT7/protamine nanocomplex was diffused and extravasated through the dense collagen matrix of the tumor. Thus, the study describes a successful application of functionalized PEG-LHT/protamine nanocomplex that can inhibit angiogenesis with long circulating, passive targeting, and tumor extravasating ability.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Heparina de Bajo-Peso-Molecular/análogos & derivados , Nanoestructuras/administración & dosificación , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Protaminas/administración & dosificación , Ácido Taurocólico/análogos & derivados , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacocinética , Inhibidores de la Angiogénesis/toxicidad , Animales , Línea Celular Tumoral , Heparina de Bajo-Peso-Molecular/administración & dosificación , Heparina de Bajo-Peso-Molecular/química , Heparina de Bajo-Peso-Molecular/farmacocinética , Heparina de Bajo-Peso-Molecular/toxicidad , Humanos , Masculino , Ratones Endogámicos C3H , Ratones Desnudos , Nanoestructuras/química , Nanoestructuras/toxicidad , Neoplasias/patología , Neovascularización Patológica/patología , Polietilenglicoles/química , Protaminas/química , Protaminas/farmacocinética , Protaminas/toxicidad , Ratas Sprague-Dawley , Ácido Taurocólico/administración & dosificación , Ácido Taurocólico/química , Ácido Taurocólico/farmacocinética , Ácido Taurocólico/toxicidad , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Biomaterials ; 35(24): 6543-52, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24816287

RESUMEN

Angiogenesis, the formation of new blood vessels, plays a pivotal role in tumor progression and for this reason angiogenesis inhibitors are an important class of therapeutics for cancer treatment. Heparin-based angiogenesis inhibitors have been newly developed as one of such classes of therapeutics and possess a great promise in the clinical context. Taurocholate conjugated low molecular weight heparin derivative (LHT7) has been proven to be a potent, multi-targeting angiogenesis inhibitor against broad-spectrum angiogenic tumors. However, major limitations of LHT7 are its poor oral bioavailability, short half-life, and frequent parenteral dosing schedule. Addressing these issues, we have developed an oral formulation of LHT7 by chemically conjugating LHT7 with a tetrameric deoxycholic acid named LHTD4, and then physically complexing it with deoxycholylethylamine (DCK). The resulting LHTD4/DCK complex showed significantly enhanced oral bioavailability (34.3 ± 2.89%) and prolonged the mean residence time (7.5 ± 0.5 h). The LHTD4/DCK complex was mostly absorbed in the intestine by transcellular pathway via its interaction with apical sodium bile acid transporter. In vitro, the VEGF-induced sprouting of endothelial spheroids was significantly blocked by LHTD4. LHTD4/DCK complex significantly regressed the total vessel fractions of tumor (77.2 ± 3.9%), as analyzed by X-ray microCT angiography, thereby inhibiting tumor growth in vivo. Using the oral route of administration, we showed that LHTD4/DCK complex could be effective and chronically administered as angiogenesis inhibitor.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Ácido Desoxicólico/farmacología , Heparina/farmacología , Administración Oral , Inhibidores de la Angiogénesis/sangre , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacocinética , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Disponibilidad Biológica , Células CACO-2 , Proliferación Celular/efectos de los fármacos , Ácido Desoxicólico/química , Heparina/química , Heparina de Bajo-Peso-Molecular/análogos & derivados , Heparina de Bajo-Peso-Molecular/síntesis química , Heparina de Bajo-Peso-Molecular/química , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Absorción Intestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Intestinos/fisiología , Masculino , Neoplasias/patología , Neovascularización Fisiológica/efectos de los fármacos , Oxidación-Reducción , Ratas Sprague-Dawley , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Ácido Taurocólico/análogos & derivados , Ácido Taurocólico/síntesis química , Ácido Taurocólico/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA