Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 193
Filtrar
1.
BMC Cancer ; 21(1): 1176, 2021 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732161

RESUMEN

BACKGROUND: According to the NAPOLI-1 trial, nanoliposomal irinotecan (nal-IRI) plus fluorouracil/folinic acid (5-FU/LV) showed improved overall survival compared to fluorouracil alone for patients with metastatic pancreatic cancer who were previously treated with gemcitabine-based therapy. In that trial, Asian patients had frequent dose modification due to haematological toxicity. There has been limited information on the clinical benefits and toxicity of this regimen in real-world settings. In this study, we assessed real-world experience of nal-IRI plus 5-FU/LV in patients with advanced pancreatic cancer after gemcitabine failure. METHODS: We conducted a single institution, retrospective analysis of response, survival and safety in patients who had been treated with nal-IRI with 5-FU/LV. Patients with metastatic pancreatic ductal adenocarcinoma previously treated with gemcitabine-based therapy received nal-IRI (80 mg/m2) with 5-FU/LV every 2 weeks. Kaplan-Meier analysis was performed to obtain median progression free survival and median overall survival. The hazard ratio and 95% confidence interval (CI) were estimated using a stratified Cox regression model. A multivariate Cox proportional hazards regression model was used to identify the effects of clinical factors. RESULTS: Fifty-one patients received nal-IRI plus 5-FU/LV between January 2015 and December 2020. The median age was 67 years, and males were 58.8%. A total of 40 (78.4%) and 11 (21.6%) patients had received one and two lines of prior chemotherapy before enrollment, respectively. Median progression-free survival was 2.8 months (95% CI 1.8-3.7) and median overall survival was 7.0 months (95% CI 6.0-7.9). Chemotherapy doses were reduced or delayed in 33 (64.7%) patients during the first 6 weeks and median relative dose intensity was 0.87. Thirty-six (70.6%) patients experienced grade 3 or 4 adverse events, most commonly neutropenia (58.8%). Most non-haematologic adverse events were under grade 2. Since the start of first-line chemotherapy, median overall survival was 16.3 months (95% CI 14.1-18.4). CONCLUSIONS: Nal-IRI plus 5-FU/LV seems to be effective, with manageable toxicities, following gemcitabine-based treatment in patients with metastatic pancreatic ductal adenocarcinoma. Nal-IRI plus 5-FU/LV following gemcitabine with nab-paclitaxel is a feasible sequential treatment option in patients with metastatic pancreatic cancer. TRIAL REGISTRATION: Retrospectively registered.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinoma Ductal Pancreático/tratamiento farmacológico , Fluorouracilo/administración & dosificación , Irinotecán/administración & dosificación , Leucovorina/administración & dosificación , Neoplasias Pancreáticas/tratamiento farmacológico , Anciano , Albúminas/uso terapéutico , Antimetabolitos Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/secundario , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Esquema de Medicación , Femenino , Fluorouracilo/efectos adversos , Humanos , Irinotecán/efectos adversos , Irinotecán/análogos & derivados , Estimación de Kaplan-Meier , Leucovorina/efectos adversos , Liposomas , Masculino , Persona de Mediana Edad , Nanoconjugados/administración & dosificación , Nanoconjugados/efectos adversos , Neutropenia/inducido químicamente , Paclitaxel/uso terapéutico , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Supervivencia sin Progresión , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Gemcitabina
2.
Nanomedicine ; 34: 102376, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33667725

RESUMEN

Alcohol abuse induces the expression of inflammatory mediators by activating the immune receptors to trigger neuroinflammation and brain damage; however, therapies that reduce neuroimmune system activation may protect against alcohol's damaging effects. Curcuminoids possess anti-inflammatory properties but suffer from low bioavailability; therefore, we designed a new receptor-targeted biodegradable star-shaped crosslinked polypeptide polymer that bears propargylamine moieties and bisdemethoxycurcumin (StClPr-BDMC-ANG) as an enhanced anti-inflammatory therapeutic that penetrates the blood-brain-barrier and ameliorates alcohol-induced neuroinflammation. StClPr-BDMC-ANG administration maintains the viability of primary glia and inhibits the ethanol-induced upregulation of crucial inflammatory mediators in the prefrontal and medial cortex in a mouse model of chronic ethanol consumption. StClPr-BDMC-ANG treatment also suppresses the ethanol-mediated downregulation of microRNAs known to negatively modulate neuroinflammation in the brain cortex (miRs 146a-5p and let-7b-5p). In summary, our results demonstrate the attenuation of alcohol-induced neuroinflammation by an optimized and targeted polypeptide-based nanoconjugate of a curcuminoid.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Curcumina/análogos & derivados , Nanoconjugados/administración & dosificación , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Péptidos/administración & dosificación , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Curcumina/administración & dosificación , Curcumina/química , Ratones , Nanoconjugados/química , Enfermedades Neuroinflamatorias/inducido químicamente , Péptidos/química
3.
Molecules ; 26(3)2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33499047

RESUMEN

Epidermal growth factor receptor (EGFR) is estimated to be overexpressed in 60~80% of colorectal cancer (CRC), which is associated with a poor prognosis. Anti-EGFR targeted monoclonal antibodies (cetuximab and panitumumab) have played an important role in the treatment of metastatic CRC. However, the therapeutic response of anti-EGFR monoclonal antibodies is limited due to multiple resistance mechanisms. With the discovery of new functions for gold nanoparticles (AuNPs), we hypothesize that cetuximab-conjugated AuNPs (cetuximab-AuNPs) will not only improve the cytotoxicity for cancer cells, but also introduce expression change of the related biomarkers on cancer cell surface. In this contribution, we investigated the size-dependent cytotoxicity of cetuximab-AuNPs to CRC cell line (HT-29), while also monitored the expression of cell surface biomarkers in response to treatment with cetuximab and cetuximab-AuNPs. AuNPs with the size of 60 nm showed the highest impact for cell cytotoxicity, which was tested by cell counting kit-8 (CCK-8) assay. Three cell surface biomarkers including epithelial cell adhesion molecule (EpCAM), melanoma cell adhesion molecule (MCAM), and human epidermal growth factor receptor-3 (HER-3) were found to be expressed at higher heterogeneity when cetuximab was conjugated to AuNPs. Both surface-enhanced Raman scattering/spectroscopy (SERS) and flow cytometry demonstrated the correlation of cell surface biomarkers in response to the drug treatment. We thus believe this study provides powerful potential for drug-conjugated AuNPs to enhance cancer prognosis and therapy.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Cetuximab/administración & dosificación , Neoplasias Colorrectales/tratamiento farmacológico , Nanopartículas del Metal/administración & dosificación , Biomarcadores de Tumor/metabolismo , Antígeno CD146/metabolismo , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Molécula de Adhesión Celular Epitelial/metabolismo , Oro , Células HT29 , Humanos , Nanopartículas del Metal/ultraestructura , Nanoconjugados/administración & dosificación , Nanoconjugados/ultraestructura , Tamaño de la Partícula , Fenotipo , Receptor ErbB-3/metabolismo , Transducción de Señal/efectos de los fármacos , Espectrometría Raman
4.
Proc Natl Acad Sci U S A ; 114(16): 4129-4134, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28373576

RESUMEN

RNA interference (RNAi)-based gene regulation platforms have shown promise as a novel class of therapeutics for the precision treatment of cancer. Techniques in preclinical evaluation of RNAi-based nanoconjugates have yet to allow for optimization of their gene regulatory activity. We have developed spherical nucleic acids (SNAs) as a blood-brain barrier-/blood-tumor barrier-penetrating nanoconjugate to deliver small interfering (si) and micro (mi)RNAs to intracranial glioblastoma (GBM) tumor sites. To identify high-activity SNA conjugates and to determine optimal SNA treatment regimens, we developed a reporter xenograft model to evaluate SNA efficacy in vivo. Engrafted tumors stably coexpress optical reporters for luciferase and a near-infrared (NIR) fluorescent protein (iRFP670), with the latter fused to the DNA repair protein O6-methylguanine-DNA-methyltransferase (MGMT). Using noninvasive imaging of animal subjects bearing reporter-modified intracranial xenografts, we quantitatively assessed MGMT knockdown by SNAs composed of MGMT-targeting siRNA duplexes (siMGMT-SNAs). We show that systemic administration of siMGMT-SNAs via single tail vein injection is capable of robust intratumoral MGMT protein knockdown in vivo, with persistent and SNA dose-dependent MGMT silencing confirmed by Western blotting of tumor tissue ex vivo. Analyses of SNA biodistribution and pharmacokinetics revealed rapid intratumoral uptake and significant intratumoral retention that increased the antitumor activity of coadministered temozolomide (TMZ). Our study demonstrates that dual noninvasive bioluminescence and NIR fluorescence imaging of cancer xenograft models represents a powerful in vivo strategy to identify RNAi-based nanotherapeutics with potent gene silencing activity and will inform additional preclinical and clinical investigations of these constructs.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Metilasas de Modificación del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Glioblastoma/tratamiento farmacológico , Nanoconjugados/administración & dosificación , ARN Interferente Pequeño/genética , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Animales , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Femenino , Fluorescencia , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones , Ratones SCID , Nanoconjugados/química , Interferencia de ARN , Temozolomida , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nano Lett ; 19(8): 5515-5523, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31362507

RESUMEN

Designing simple-structured nanomedicine without lacking key functionalities, thereby avoiding incomplete damage or relapse of tumor with the administration of a safe dose, is pivotal for successful cancer nanotherapy. We herein presented a nanomedicine of photodynamic therapy (PDT) that simply assembled amphiphilic macromolecules of poly-l-lysine conjugating with photosensitizers onto hydrophobic upconverting nanoparticles. We demonstrated that the nanoformulation, despite its simple structure and synthesis, simultaneously possesses multiple features, including substantial payload of photosensitizers, avid cellular internalization both in vitro and in vivo, efficient diffusion and broad distribution in tumor lesion, and potent fatality for cancer stem cells that are refractory to other therapy modalities. Because of the combination of these functionalities, the tumors in mice were eradicated and no relapse was observed after at least 40 days, just with an extremely low intraperitoneal injection dose of 5.6 mg/kg. Our results suggested a strategy for designing multifunctional nanomedicines with simple construct and efficacious therapeutic response and presented the promising potential of PDT for a radical cure of cancer.


Asunto(s)
Nanoconjugados/uso terapéutico , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico , Animales , Línea Celular Tumoral , Células HeLa , Humanos , Ratones , Nanoconjugados/administración & dosificación , Nanoconjugados/química , Recurrencia Local de Neoplasia/prevención & control , Células Madre Neoplásicas/efectos de los fármacos , Fotoquimioterapia , Fármacos Fotosensibilizantes/administración & dosificación , Fármacos Fotosensibilizantes/química , Polilisina/administración & dosificación , Polilisina/análogos & derivados , Polilisina/uso terapéutico
6.
Nano Lett ; 19(3): 1719-1727, 2019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30724087

RESUMEN

Protein nanocages are promising multifunctional platforms for nanomedicine owing to the ability to decorate their surfaces with multiple functionalities through genetic and/or chemical modification to achieve desired properties for therapeutic and diagnostic purposes. Here, we describe a model antigen (OVA peptide) that was conjugated to the surface of a naturally occurring hepatitis B core protein nanocage (HBc NC) by genetic modification. The engineered OVA-HBc nanocages (OVA-HBc NCs), displaying high density repetitive array of epitopes in a limited space by self-assembling into symmetrical structure, not only can induce bone marrow derived dendritic cells (BMDC) maturation effectively but also can be enriched in the draining lymph nodes. Naïve C57BL/6 mice immunized with OVA-HBc NCs are able to generate significant and specific cytotoxic T lymphocyte (CTL) responses. Moreover, OVA-HBc NCs as a robust nanovaccine can trigger preventive antitumor immunity and significantly delay tumor growth. When combined with a low-dose chemotherapy drug (paclitaxel), OVA-HBc NCs could specifically inhibit progression of an established tumor. Our findings support HBc-based nanocages with modularity and scalability as an attractive nanoplatform for combination cancer immunotherapy.


Asunto(s)
Antígenos de Neoplasias/administración & dosificación , Antígenos del Núcleo de la Hepatitis B/inmunología , Nanoconjugados/administración & dosificación , Neoplasias/terapia , Animales , Antígenos de Neoplasias/inmunología , Bioingeniería/métodos , Proliferación Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Epítopos/genética , Epítopos/inmunología , Hepatitis B/inmunología , Antígenos del Núcleo de la Hepatitis B/administración & dosificación , Humanos , Inmunoterapia/métodos , Ratones Endogámicos C57BL , Neoplasias/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
7.
Nano Lett ; 19(11): 7573-7587, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31518145

RESUMEN

Despite untiring efforts to develop therapies for pancreatic ductal adenocarcinoma (PDAC), survival statistics remain dismal, necessitating distinct approaches. Photodynamic priming (PDP), which improves drug delivery and combination regimens, as well as tumor photodestruction are key attributes of photodynamic therapy (PDT), making it a distinctive clinical option for PDAC. Localized, high-payload nanomedicine-assisted delivery of photosensitizers (PSs), with molecular specificity and controlled photoactivation, thus becomes critical in order to reduce collateral toxicity during more expansive photodynamic activation procedures with curative intent. As such, targeted photoactivable lipid-based nanomedicines are an ideal candidate but have failed to provide greater than two-fold cancer cell selectivity, if at all, due to their extensive multivariant physical, optical, and chemical complexity. Here, we report (1) a systematic multivariant tuning approach to engineer (Cet, anti-EGFR mAb) photoimmunonanoconjugates (PINs), and (2) stroma-rich heterotypic PDAC in vitro and in vivo models incorporating patient-derived pancreatic cancer-associated fibroblasts (PCAFs) that recapitulate the desmoplasia observed in the clinic. These offer a comprehensive, disease-specific framework for the development of Cet-PINs. Specificity-tuning of the PINs, in terms of PS lipid anchoring, electrostatic modulation, Cet orientation, and Cet surface densities, achieved ∼16-fold binding specificities and rapid penetration of the heterotypic organoids within 1 h, thereby providing a ∼16-fold enhancement in molecular targeted NIR photodestruction. As a demonstration of their inherent amenability for multifunctionality, encapsulation of high payloads of gemcitabine hydrochloride, 5-fluorouracil, and oxaliplatin within the Cet-PINs further improved their antitumor efficacy in the heterotypic organoids. In heterotypic desmoplastic tumors, the Cet-PINs efficiently penetrated up to 470 µm away from blood vessels, and photodynamic activation resulted in substantial tumor necrosis, which was not elicited in T47D tumors (low EGFR) or when using untargeted constructs in both tumor types. Photodynamic activation of the Cet-PINs in the heterotypic desmoplastic tumors resulted in collagen photomodulation, with a 1.5-fold reduction in collagen density, suggesting that PDP may also hold potential for conquering desmoplasia. The in vivo safety profile of photodynamic activation of the Cet-PINs was also substantially improved, as compared to the untargeted constructs. While treatment using the Cet-PINs did not cause any detriment to the mice's health or to healthy proximal tissue, photodynamic activation of untargeted constructs induced severe acute cachexia and weight loss in all treated mice, with substantial peripheral skin necrosis, muscle necrosis, and bowel perforation. This study is the first report demonstrating the true value of molecular targeting for NIR-activable PINs. These constructs integrate high payload delivery, efficient photodestruction, molecular precision, and collagen photomodulation in desmoplastic PDAC tumors in a single treatment using a single construct. Such combined PIN platforms and heterocellular models open up an array of further multiplexed combination therapies to synergistically control desmoplastic tumor progression and extend PDAC patient survival.


Asunto(s)
Carcinoma Ductal Pancreático/tratamiento farmacológico , Inmunoconjugados/uso terapéutico , Nanoconjugados/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico , Animales , Anticuerpos Monoclonales/uso terapéutico , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/patología , Carcinoma Ductal Pancreático/patología , Sistemas de Liberación de Medicamentos/métodos , Receptores ErbB/antagonistas & inhibidores , Humanos , Inmunoconjugados/administración & dosificación , Ratones , Nanoconjugados/administración & dosificación , Nanomedicina/métodos , Organoides/efectos de los fármacos , Organoides/patología , Neoplasias Pancreáticas/patología , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/administración & dosificación
8.
Pak J Pharm Sci ; 33(2): 705-714, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32276917

RESUMEN

Human diseases like viral organisms for example, hepatitis, HIV and etc., attack the health and caused large mortality in populations by many years. So finding novel delivery vehicles based antiviral drugs employing nano-materials is of high universal interest. In current approach a very biocompatible biodegradable nano-biopolymer anionic linear globular dendrimer second generation G2 was elaborately conjugated to a well-known anti-HIV drug Azidovudine and thereafter was characterized by different analytical techniques like AFM, Zeta sizer, 1HNMR, FTIR and LC-Mass spectroscopy. Then, Anionic Linear Globular DendrimerG2-Zidovudine Nano-Conjugate was assessed on human normal cells (toxicity assay by XTT test) and also HIV cell model and the results showed that Anionic Linear Globular DendrimerG2-Zidovudine Nano-Conjugate Significantly Decreased Retroviral Activity without any human cell toxicity respectively. Based on current experimental data such nano-compositions is proposed for further in vivo anti-HIV assays as well.


Asunto(s)
Antirretrovirales/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Dendrímeros/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Nanoconjugados/administración & dosificación , Zidovudina/administración & dosificación , Aniones , Antirretrovirales/química , Supervivencia Celular/fisiología , Dendrímeros/química , Relación Dosis-Respuesta a Droga , Células HEK293 , VIH-1/efectos de los fármacos , VIH-1/fisiología , Humanos , Nanoconjugados/química , Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Estearatos/administración & dosificación , Estearatos/química , Zidovudina/química
9.
Proc Natl Acad Sci U S A ; 113(32): E4601-9, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27457945

RESUMEN

Malignant osteolysis associated with inoperable primary bone tumors and multifocal skeletal metastases remains a challenging clinical problem in cancer patients. Nanomedicine that is able to target and deliver therapeutic agents to diseased bone sites could potentially provide an effective treatment option for different types of skeletal cancers. Here, we report the development of polylactide nanoparticles (NPs) loaded with doxorubicin (Doxo) and coated with bone-seeking pamidronate (Pam) for the targeted treatment of malignant skeletal tumors. In vivo biodistribution of radiolabeled targeted Pam-NPs demonstrated enhanced bone tumor accumulation and prolonged retention compared with nontargeted NPs. In a murine model of focal malignant osteolysis, Pam-functionalized, Doxo-loaded NPs (Pam-Doxo-NPs) significantly attenuated localized osteosarcoma (OS) progression compared with nontargeted Doxo-NPs. Importantly, we report on the first evaluation to our knowlege of Pam-Doxo-NPs in dogs with OS, which possess tumors of anatomic size and physiology comparable to those in humans. The repeat dosing of Pam-Doxo-NPs in dogs with naturally occurring OS indicated the therapeutic was well tolerated without hematologic, nonhematologic, and cardiac toxicities. By nuclear scintigraphy, the biodistribution of Pam-Doxo-NPs demonstrated malignant bone-targeting capability and exerted measurable anticancer activities as confirmed with percent tumor necrosis histopathology assessment.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Óseas/tratamiento farmacológico , Difosfonatos/administración & dosificación , Doxorrubicina/administración & dosificación , Nanoconjugados/administración & dosificación , Osteólisis/tratamiento farmacológico , Animales , Difosfonatos/farmacocinética , Doxorrubicina/toxicidad , Femenino , Ratones , Ratones Endogámicos BALB C , Pamidronato
10.
Drug Dev Ind Pharm ; 45(9): 1496-1505, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31241372

RESUMEN

Nanomaterial based anticancer treatment is promising nowadays because of their small size that can penetrate and interact both inside and outside the cell surface. In this study, a simple protocol was followed for the conjugation of the biologically synthesized selenium nanoparticles (SeNPs) and short chain synthetic peptide. SeNPs was synthesized by using the culture supernatant of Streptomyces griseoruber, actinomycetes isolated from the soil. The short chain peptide Boc-L-F-OMe was synthesized by the conventional solution phase chemistry using a racemization-free fragment condensation strategy. Peptide interaction with different anticancer receptors was preliminarily studied by docking studies. Biosynthesized SeNPs was conjugated with short chain synthetic peptides by means of cysteine conjugation. Characterization of SeNPs with peptide was done by UV-visible spectroscopy and DLS that showed the red shift in the peak and increase in average particle size and zeta potential, respectively. Bioconjugated SeNPs- peptide was tested for its cytotoxicity against the colon cancer cell line HT-29. Bioconjugated SeNPs-peptide showed enhanced cytotoxic activity when compared to the peptide and nanoparticle alone that was tested at 10-50 µg/ml concentration. Further apoptotic studies were done by AO/PI staining and DNA fragmentation assay that confirms the cytotoxicity of the conjugates. Novel peptide-SeNPs conjugates tested in our study has a significant anticancer activity that can be potentially used for targeting the cancer cells.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/administración & dosificación , Neoplasias Colorrectales/tratamiento farmacológico , Nanoconjugados/administración & dosificación , Adenocarcinoma/patología , Antineoplásicos/química , Neoplasias Colorrectales/patología , Dipéptidos/administración & dosificación , Evaluación Preclínica de Medicamentos , Células HT29 , Humanos , Simulación del Acoplamiento Molecular , Nanoconjugados/química , Selenio
11.
J Allergy Clin Immunol ; 141(6): 2121-2131, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29655584

RESUMEN

BACKGROUND: Immunotherapy for food allergies involves progressive increased exposures to food that result in desensitization to food allergens in some subjects but not tolerance to the food. Therefore new approaches to suppress allergic immunity to food are necessary. Previously, we demonstrated that intranasal immunization with a nanoemulsion (NE) adjuvant induces robust mucosal antibody and TH17-polarized immunity, as well as systemic TH1-biased cellular immunity with suppression of pre-existing TH2-biased immunity. OBJECTIVE: We hypothesized that immunization with food in conjunction with the nanoemulsion adjuvant could lead to modulation of allergic reactions in food allergy by altering pre-existing allergic immunity and enhancing mucosal immunity. METHODS: Mice were sensitized to peanut with aluminum hydroxide or cholera toxin. The animals were then administered 3 monthly intranasal immunizations with peanut in the nanoemulsion adjuvant or saline. Mice were then challenged with peanut to examine allergen reactivity. RESULTS: The NE intranasal immunizations resulted in marked decreases in TH2 cytokine, IgG1, and IgE levels, whereas TH1 and mucosal TH17 immune responses were increased. After allergen challenge, these mice showed significant reductions in allergic hypersensitivity. Additionally, the NE immunizations significantly increased antigen-specific IL-10 production and regulatory T-cell counts, and the protection induced by NE was dependent in part on IL-10. Control animals immunized with intranasal peanut in saline had no modulation of their allergic response. CONCLUSIONS: NE adjuvant-mediated induction of mucosal TH17 and systemic TH1-biased immunity can suppress TH2-mediated allergy through multiple mechanisms and protect against anaphylaxis. These results suggest the potential therapeutic utility of this approach in the setting of food allergy.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Desensibilización Inmunológica/métodos , Hipersensibilidad al Cacahuete/inmunología , Células Th2/inmunología , Administración Intranasal , Animales , Modelos Animales de Enfermedad , Emulsiones , Femenino , Ratones , Nanoconjugados/administración & dosificación , Células Th2/efectos de los fármacos
12.
Bioconjug Chem ; 29(3): 813-823, 2018 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-29148731

RESUMEN

Current strategies for treating autoimmunity involve the administration of broad-acting immunosuppressive agents that impair healthy immunity. Intravenous (i.v.) administration of poly(lactide- co-glycolide) nanoparticles (NPs) containing disease-relevant antigens (Ag-NPs) have demonstrated antigen (Ag)-specific immune tolerance in models of autoimmunity. However, subcutaneous (s.c.) delivery of Ag-NPs has not been effective. This investigation tested the hypothesis that codelivery of the immunomodulatory cytokine, transforming growth factor beta 1 (TGF-ß), on Ag-NPs would modulate the immune response to Ag-NPs and improve the efficiency of tolerance induction. TGF-ß was coupled to the surface of Ag-NPs such that the loadings of Ag and TGF-ß were independently tunable. The particles demonstrated bioactive delivery of Ag and TGF-ß in vitro by reducing the inflammatory phenotype of bone marrow-derived dendritic cells and inducing regulatory T cells in a coculture system. Using an in vivo mouse model for multiple sclerosis, experimental autoimmune encephalomyelitis, TGF-ß codelivery on Ag-NPs resulted in improved efficacy at lower doses by i.v. administration and significantly reduced disease severity by s.c. administration. This study demonstrates that the codelivery of immunomodulatory cytokines on Ag-NPs may enhance the efficacy of Ag-specific tolerance therapies by programming Ag presenting cells for more efficient tolerance induction.


Asunto(s)
Antígenos/administración & dosificación , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Factores Inmunológicos/administración & dosificación , Esclerosis Múltiple/tratamiento farmacológico , Nanoconjugados/administración & dosificación , Poliglactina 910/administración & dosificación , Factor de Crecimiento Transformador beta/administración & dosificación , Animales , Antígenos/química , Antígenos/uso terapéutico , Células Cultivadas , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Tolerancia Inmunológica/efectos de los fármacos , Factores Inmunológicos/química , Factores Inmunológicos/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Nanoconjugados/química , Nanoconjugados/uso terapéutico , Poliglactina 910/química , Poliglactina 910/uso terapéutico , Factor de Crecimiento Transformador beta/química , Factor de Crecimiento Transformador beta/uso terapéutico
13.
Mol Pharm ; 15(12): 5754-5761, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30376341

RESUMEN

Ligand-decorated nanoparticles are extensively studied and applied for in vivo drug delivery and molecular imaging. Generally, two different ligand-decoration procedures are utilized; ligands are either conjugated with nanoparticle ingredients and incorporated during nanoparticle preparation, or they are attached to preformed nanoparticles by utilizing functionalized reactive surface groups (e.g., maleimide). Although the two procedures result in nanoparticles with very similar physicochemical properties, formulations obtained through the latter manufacturing process typically contain nonconjugated reactive surface groups. In the current study, we hypothesized that the different ligand-decoration procedures might affect the extent of interaction between nanoparticles and immune cells (especially phagocytes). In order to investigate our hypothesis, we decorated lipidic nanoparticles with a widely used cyclic Arg-Gly-Asp (cRGD) peptide using the two different procedures. As proven from in vivo experiments in mice, the presence of nonconjugated surface moieties results in increased recognition by the immune system. This is important knowledge considering the emerging focus on understanding and optimizing ways to target and track immune cells and the development of nanomedicine-based strategies in the field of immunotherapy.


Asunto(s)
Composición de Medicamentos/métodos , Nanoconjugados/administración & dosificación , Oligopéptidos/administración & dosificación , Fagocitos/efectos de los fármacos , Animales , Evaluación Preclínica de Medicamentos , Inmunoterapia/métodos , Ligandos , Liposomas , Maleimidas/química , Ratones , Ratones Endogámicos BALB C , Nanoconjugados/química , Nanomedicina/métodos , Oligopéptidos/química , Fagocitos/inmunología
14.
J Neurosci ; 36(12): 3441-52, 2016 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-27013674

RESUMEN

Respiratory complications in patients with spinal cord injury (SCI) are common and have a negative impact on the quality of patients' lives. Systemic administration of drugs that improve respiratory function often cause deleterious side effects. The present study examines the applicability of a novel nanotechnology-based drug delivery system, which induces recovery of diaphragm function after SCI in the adult rat model. We developed a protein-coupled nanoconjugate to selectively deliver by transsynaptic transport small therapeutic amounts of an A1 adenosine receptor antagonist to the respiratory centers. A single administration of the nanoconjugate restored 75% of the respiratory drive at 0.1% of the systemic therapeutic drug dose. The reduction of the systemic dose may obviate the side effects. The recovery lasted for 4 weeks (the longest period studied). These findings have translational implications for patients with respiratory dysfunction after SCI. SIGNIFICANCE STATEMENT: The leading causes of death in humans following SCI are respiratory complications secondary to paralysis of respiratory muscles. Systemic administration of methylxantines improves respiratory function but also leads to the development of deleterious side effects due to actions of the drug on nonrespiratory sites. The importance of the present study lies in the novel drug delivery approach that uses nanotechnology to selectively deliver recovery-inducing drugs to the respiratory centers exclusively. This strategy allows for a reduction in the therapeutic drug dose, which may reduce harmful side effects and markedly improve the quality of life for SCI patients.


Asunto(s)
Diafragma/fisiopatología , Receptor de Adenosina A1/metabolismo , Parálisis Respiratoria/tratamiento farmacológico , Parálisis Respiratoria/fisiopatología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/fisiopatología , Xantinas/administración & dosificación , Antagonistas del Receptor de Adenosina A1/administración & dosificación , Antagonistas del Receptor de Adenosina A1/química , Animales , Diafragma/efectos de los fármacos , Masculino , Fuerza Muscular/efectos de los fármacos , Nanoconjugados/administración & dosificación , Nanoconjugados/química , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Mecánica Respiratoria/efectos de los fármacos , Parálisis Respiratoria/etiología , Traumatismos de la Médula Espinal/complicaciones , Resultado del Tratamiento , Aglutinina del Germen de Trigo-Peroxidasa de Rábano Silvestre Conjugada/química , Aglutinina del Germen de Trigo-Peroxidasa de Rábano Silvestre Conjugada/farmacocinética , Xantinas/química
15.
Small ; 12(19): 2595-608, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27027489

RESUMEN

The circulating tumor cells (CTCs) existing in cancer survivors are considered the root cause of cancer metastasis. To prevent the devastating metastasis cascade from initiation, we hypothesize that a biodegradable nanomaterial loaded with the abortifacient mifepristone (MIF) and conjugated with the epithelial cell adhesion molecule antibody (aEpCAM) may serve as a safe and effective cancer metastatic preventive agent by targeting CTCs and preventing their adhesion-invasion to vascular intima. It is demonstrated that MIF-loaded mesoporous silica nanoparticles (MSN) coated with aEpCAM (aE-MSN-M) can specifically target and bind colorectal cancer cells in either cell medium or blood through EpCAM recognition proven by quantitative flow cytometric detection and free aEpCAM competitive assay. The specific binding results in downregulation of the captured cells and drives them into G0/G1 phase primarily attributed to the effect of aEpCAM. The functional nanoparticles significantly inhibit the heteroadhesion between cancer cells and endothelial cells, suggesting the combined inhibition effects of aEpCAM and MIF on E-selectin and ICAM-1 expression. The functionalized nanoparticles circulate in mouse blood long enough to deliver MIF and inhibit lung metastasis. The present proof-of-concept study shows that the aE-MSN-M can prevent cancer metastasis by restraining CTC activity and their adhesion-invasion to vascular intima.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/secundario , Molécula de Adhesión Celular Epitelial/inmunología , Mifepristona/administración & dosificación , Nanocápsulas/química , Dióxido de Silicio/química , Abortivos Esteroideos/administración & dosificación , Abortivos Esteroideos/química , Absorción Fisicoquímica , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Apoptosis/efectos de los fármacos , Neoplasias Colorrectales/inmunología , Difusión , Sistemas de Liberación de Medicamentos/métodos , Reposicionamiento de Medicamentos , Células HT29 , Humanos , Ratones , Mifepristona/química , Nanocápsulas/ultraestructura , Nanoconjugados/administración & dosificación , Nanoconjugados/química , Nanoporos/ultraestructura , Resultado del Tratamiento
16.
Bioconjug Chem ; 27(12): 2874-2885, 2016 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-27998075

RESUMEN

The synthesis and anticancer activity of a copper(II) diacetyl-bis(N4-methylthiosemicarbazone) complex and its nanoconjugates are reported. The copper(II) complex is connected to a carboxylic acid group through a cleavable disulfide link to enable smart delivery. The copper complex is tethered to highly water-soluble 20 nm gold nanoparticles (AuNPs), stabilized by amine terminated lipoic acid-polyethylene glycol (PEG). The gold nanoparticle carrier was further decorated with biotin to achieve targeted action. The copper complex and the conjugates with and without biotin, were tested against HeLa and HaCaT cells. They show very good anticancer activity against HeLa cells, a cell line derived from cervical cancer and are less active against HaCaT cells. Slow and sustained release of the complex from conjugates is demonstrated through cleavage of disulfide linker in the presence of glutathione (GSH), a reducing agent intrinsically present in high concentrations within cancer cells. Biotin appended conjugates do not show greater activity than conjugates without biotin against HeLa cells. This is consistent with drug uptake studies, which suggests similar uptake profiles for both conjugates in vitro. However, in vivo studies using a HeLa cell xenograft tumor model shows 3.8-fold reduction in tumor volume for the biotin conjugated nanoparticle compared to the control whereas the conjugate without biotin shows only 2.3-fold reduction in the tumor volume suggesting significant targeting.


Asunto(s)
Antineoplásicos/administración & dosificación , Cobre/química , Sistemas de Liberación de Medicamentos/métodos , Nanoconjugados/administración & dosificación , Nanopartículas/química , Animales , Antineoplásicos/química , Biotina/química , Técnicas de Química Sintética , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Cobre/administración & dosificación , Femenino , Oro/química , Humanos , Ratones Desnudos , Nanoconjugados/química , Polietilenglicoles/química , Ácido Tióctico/química , Tiosemicarbazonas/química , Ensayos Antitumor por Modelo de Xenoinjerto
17.
NMR Biomed ; 29(10): 1436-44, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27552925

RESUMEN

In many human diseases, the presence of inflammation is associated with an increase in the level of reactive oxygen species (ROS). The resulting state of oxidative stress is highly detrimental and can initiate a cascade of events that ultimately lead to cell death. Thus, many therapeutic attempts have been focused on either modulating the immune system to lower inflammation or reducing the damaging caused by ROS. Berlin et al. reported the development of a novel nanoantioxidant known as poly(ethylene glycol)-functionalized-hydrophilic carbon clusters (PEG-HCCs). They showed that PEG-HCCs could be targeted to cancer cells, utilized as a drug delivery vector, and can even be visualized ex vivo. Our work here furthers this work and characterizes Gd-DTPA conjugated PEG-HCCs and explores the potential for in vivo tracking of T cells in live mice. We utilized a mouse model of delayed-type hypersensitivity (DTH) to assess the immunomodulatory effects of PEG-HCCs. The T1 -agent Gd-DTPA was then conjugated to the PEG-HCCs and T1 measurements, and T1 -weighted MRI of the modified PEG-HCCs was done to assess their relaxivity. We then assessed if PEG-HCCs could be visualized both ex vivo and in vivo within the mouse lymph node and spleen. Mice treated with PEG-HCCs showed significant improvements in the DTH assay as compared to the vehicle (saline)-treated control. Flow cytometry demonstrated that splenic T cells are capable of internalizing PEG-HCCs whereas fluorescent immunohistochemistry showed that PEG-HCCs are detectable within the cortex of lymph nodes. Finally, our nanoantioxidants can be visualized in vivo within the lymph nodes and spleen of a mouse after addition of the Gd-DTPA. PEG-HCCs are internalized by T cells in the spleen and can reduce inflammation by suppression of a recall immune response. PEG-HCCs can be modified to allow for both in vitro and in vivo visualization using MRI. © 2016 The Authors. NMR in Biomedicine published by John Wiley & Sons Ltd.


Asunto(s)
Antioxidantes/administración & dosificación , Inmunidad Innata/inmunología , Memoria Inmunológica/inmunología , Imagen por Resonancia Magnética/métodos , Nanopartículas/administración & dosificación , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Animales , Antioxidantes/química , Rastreo Celular/métodos , Células Cultivadas , Femenino , Gadolinio DTPA/química , Inmunidad Innata/efectos de los fármacos , Memoria Inmunológica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Nanocápsulas/administración & dosificación , Nanocápsulas/química , Nanoconjugados/administración & dosificación , Nanoconjugados/química , Nanopartículas/química , Especies Reactivas de Oxígeno/inmunología , Linfocitos T/citología
18.
J Nanosci Nanotechnol ; 16(5): 4762-70, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27483820

RESUMEN

Gold nanoparticles (GNPs) are synthesized using the medicinal plant Leucas Aspera extract (LAE) and poly lactic acid-co-poly ethylene glycol-co-poly lactic acid (PLA-PEG-PLA) copolymer by water-in-oil (W/O) emulsion method. The proposed method of W/O emulsion technique involves synthesis of GNPs and loading of Leucas Aspera extract on to the PLA-PEG-PLA copolymer matrix simultaneously. The synthesized GNPs are characterized by Fourier transform infra-red (FTIR) spectroscopy, dynamic light scattering (DLS), X-ray diffractometry (XRD) and transmission electron microscopy (TEM). The GNPs-LAE loaded polymer NPs are examined for the in vitro cytotoxicity on South African green monkey's kidney cells. The GNPs-LAE loaded polymer nanoconjugates exhibit maximum up to 95% of cell viability with 100 µg concentration of GNPs in the sample. The GNPs-LAE loaded polymer NPs exhibit better anti-inflammatory activity when compared to the pure LAE.


Asunto(s)
Sangre/inmunología , Medicamentos Herbarios Chinos/química , Oro/toxicidad , Lactatos/química , Nanopartículas del Metal/toxicidad , Nanoconjugados/toxicidad , Polietilenglicoles/química , Absorción Fisicoquímica , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/síntesis química , Sangre/efectos de los fármacos , Chlorocebus aethiops , Difusión , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/toxicidad , Oro/administración & dosificación , Nanopartículas del Metal/administración & dosificación , Nanocápsulas , Nanoconjugados/administración & dosificación , Nanoconjugados/química , Tensoactivos/química , Células Vero
19.
Proc Natl Acad Sci U S A ; 110(37): 15127-32, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-23980155

RESUMEN

Nanoparticles are currently being investigated in a number of human clinical trials. As information on how nanoparticles function in humans is difficult to obtain, animal studies that can be correlative to human behavior are needed to provide guidance for human clinical trials. Here, we report correlative studies on animals and humans for CRLX101, a 20- to 30-nm-diameter, multifunctional, polymeric nanoparticle containing camptothecin (CPT). CRLX101 is currently in phase 2 clinical trials, and human data from several of the clinical investigations are compared with results from multispecies animal studies. The pharmacokinetics of polymer-conjugated CPT (indicative of the CRLX101 nanoparticles) in mice, rats, dogs, and humans reveal that the area under the curve scales linearly with milligrams of CPT per square meter for all species. Plasma concentrations of unconjugated CPT released from CRLX101 in animals and humans are consistent with each other after accounting for differences in serum albumin binding of CPT. Urinary excretion of polymer-conjugated CPT occurs primarily within the initial 24 h after dosing in animals and humans. The urinary excretion dynamics of polymer-conjugated and unconjugated CPT appear similar between animals and humans. CRLX101 accumulates into solid tumors and releases CPT over a period of several days to give inhibition of its target in animal xenograft models of cancer and in the tumors of humans. Taken in total, the evidence provided from animal models on the CRLX101 mechanism of action suggests that the behavior of CRLX101 in animals is translatable to humans.


Asunto(s)
Camptotecina/administración & dosificación , Ciclodextrinas/administración & dosificación , Nanoconjugados/administración & dosificación , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/uso terapéutico , Área Bajo la Curva , Camptotecina/farmacocinética , Camptotecina/uso terapéutico , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Ciclodextrinas/farmacocinética , Ciclodextrinas/uso terapéutico , Perros , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Ratones , Ratones Desnudos , Nanoconjugados/química , Nanoconjugados/uso terapéutico , Ratas , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Investigación Biomédica Traslacional
20.
Chem Soc Rev ; 44(19): 6670-83, 2015 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-26118960

RESUMEN

The advances in genomics, proteomics, and bioinformatics have directed the development of new anticancer agents to reduce drug abuse and increase safe and specific drug treatment. Theranostics, combining therapy and diagnosis, is an appealing approach for chemotherapy in medicine which exhibits improved biodistribution, selective cancer targeting ability, reduced toxicity, masked drug efficacy, and minimum side effects. The role of diagnosis tools in theranostics is to collect the information of the diseased state before and after specific treatment. Magnetic particle-, mesoporous silica-, various carbon allotrope-, and polymer nanoparticle-based theranostic systems are well accepted and clinically significant. Currently, small conjugate-based systems have received much attention for cancer treatment and diagnosis. The structural architecture of these systems is relatively simple, compact, biocompatible, and unidirectional. In this tutorial review, we summarize the latest developments on small conjugate based theranostic agents for tumor treatment and diagnosis using fluorescence and magnetic resonance imaging (MRI).


Asunto(s)
Antineoplásicos/uso terapéutico , Portadores de Fármacos , Nanoconjugados/uso terapéutico , Neoplasias , Profármacos/uso terapéutico , Nanomedicina Teranóstica/métodos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Humanos , Imagen por Resonancia Magnética , Estructura Molecular , Nanoconjugados/administración & dosificación , Nanoconjugados/química , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Profármacos/administración & dosificación , Profármacos/química
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda