Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Pharm ; 17(7): 2518-2531, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32421341

RESUMEN

M2-like tumor-associated macrophages (M2 TAMs) play important roles in the resistance of tumors to immunotherapies. Selective depletion or reprogramming of M2 TAMs may sensitize the nonresponsive tumors for immune-mediated eradication. However, precision delivery of payloads to M2 TAMs, while sparing healthy tissues, has remained an unresolved challenge. Here, we studied the application of a short linear peptide (CSPGAK, "mUNO") for the delivery of molecular and nanoscale cargoes in M2 TAMs in vitro and the relevance of the peptide for in vivo targeting of early-stage primary breast tumors and metastatic lung foci. First, we performed in silico modeling and found that mUNO interacts with mouse CD206 via a binding site between lectin domains CTLD1 and CTLD2, the same site previously demonstrated to be involved in mUNO binding to human CD206. Second, we showed that cultured M2 macrophages take up fluorescein-labeled (FAM) polymersomes conjugated with mUNO using the sulfhydryl group of its N-terminal cysteine. Pulse/chase studies of FAM-mUNO in M2 macrophages suggested that the peptide avoided lysosomal entrapment and escaped from early endosomes. Third, our in vivo studies with FAM-mUNO demonstrated that intraperitoneal administration results in better pharmacokinetics and higher blood bioavailability than can be achieved with intravenous administration. Intraperitoneal FAM-mUNO, but not FAM-control, showed a robust accumulation in M2-skewed macrophages in mouse models of early primary breast tumor and lung metastasis. This targeting was specific, as no uptake was observed in nonmalignant control organs, including the liver, or other cell types in the tumor, including M1 macrophages. Collectively, our studies support the application of the CD206-binding mUNO peptide for delivery of molecular and nanoscale cargoes to M2 macrophages and manifest the relevance of this mode of targeting primary and metastatic breast tumors.


Asunto(s)
Inmunoterapia/métodos , Lectinas Tipo C/química , Neoplasias Pulmonares/diagnóstico , Metástasis Linfática/diagnóstico , Lectinas de Unión a Manosa/química , Péptidos/química , Receptores de Superficie Celular/química , Neoplasias de la Mama Triple Negativas/diagnóstico , Macrófagos Asociados a Tumores/inmunología , Animales , Sitios de Unión , Diferenciación Celular , Línea Celular Tumoral , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacocinética , Femenino , Fluorescencia , Humanos , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Metástasis Linfática/diagnóstico por imagen , Metástasis Linfática/inmunología , Lisosomas/metabolismo , Maleimidas/química , Receptor de Manosa , Lectinas de Unión a Manosa/inmunología , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Péptidos/administración & dosificación , Péptidos/metabolismo , Péptidos/farmacocinética , Poliésteres/química , Polietilenglicoles/química , Polímeros/administración & dosificación , Polímeros/química , Polímeros/farmacología , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Neoplasias de la Mama Triple Negativas/patología , Macrófagos Asociados a Tumores/metabolismo
2.
Bioorg Med Chem Lett ; 26(6): 1618-1623, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26874401

RESUMEN

Because nucleic acids (NAs) have immense potential value as therapeutics, the development of safe and effective synthetic NA vectors continues to attract much attention. In vivo applications of NA vectors require stabilized, nanometer-scale particles, but the commonly used approaches of steric stabilization with a polymer coat (e.g., PEGylation; PEG=poly(ethylene glycol)) interfere with attachment to cells, uptake, and endosomal escape. Conjugation of peptides to PEG-lipids can improve cell attachment and uptake for cationic liposome-DNA (CL-DNA) complexes. We present several synthetic approaches to peptide-PEG-lipids and discuss their merits and drawbacks. A lipid-PEG-amine building block served as the common key intermediate in all synthetic routes. Assembling the entire peptide-PEG-lipid by manual solid phase peptide synthesis (employing a lipid-PEG-carboxylic acid) allowed gram-scale synthesis but is mostly applicable to linear peptides connected via their N-terminus. Conjugation via thiol-maleimide or strain-promoted (copper-free) azide-alkyne cycloaddition chemistry is highly amenable to on-demand preparation of peptide-PEG-lipids, and the appropriate PEG-lipid precursors are available in a single chemical step from the lipid-PEG-amine building block. Azide-alkyne cycloaddition is especially suitable for disulfide-bridged peptides such as iRGD (cyclic CRGDKGPDC). Added at 10 mol% of a cationic/neutral lipid mixture, the peptide-PEG-lipids stabilize the size of CL-DNA complexes. They also affect cell attachment and uptake of nanoparticles in a peptide-dependent manner, thereby providing a platform for preparing stabilized, affinity-targeted CL-DNA nanoparticles.


Asunto(s)
ADN/química , Lípidos/química , Liposomas/química , Péptidos Cíclicos/síntesis química , Polietilenglicoles/química , Cationes/química , Humanos , Liposomas/síntesis química , Estructura Molecular , Nanopartículas/química , Péptidos Cíclicos/química
3.
Nat Mater ; 13(9): 904-11, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24907927

RESUMEN

There is considerable interest in using nanoparticles as labels or to deliver drugs and other bioactive compounds to cells in vitro and in vivo. Fluorescent imaging, commonly used to study internalization and subcellular localization of nanoparticles, does not allow unequivocal distinction between cell surface-bound and internalized particles, as there is no methodology to turn particles 'off'. We have developed a simple technique to rapidly remove silver nanoparticles outside living cells, leaving only the internalized pool for imaging or quantification. The silver nanoparticle (AgNP) etching is based on the sensitivity of Ag to a hexacyanoferrate-thiosulphate redox-based destain solution. In demonstration of the technique we present a class of multicoloured plasmonic nanoprobes comprising dye-labelled AgNPs that are exceptionally bright and photostable, carry peptides as model targeting ligands, can be etched rapidly and with minimal toxicity in mice, and that show tumour uptake in vivo.


Asunto(s)
Células/metabolismo , Nanopartículas del Metal , Imagen Molecular/métodos , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Plata/química , Plata/metabolismo , Animales , Avidina/química , Transporte Biológico , Línea Celular Tumoral , Femenino , Humanos , Ratones , Sondas Moleculares/análisis , Sondas Moleculares/toxicidad , Polietilenglicoles/química , Plata/toxicidad
4.
ACS Nano ; 18(22): 13983-13999, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38767983

RESUMEN

In recent years, steady progress has been made in synthesizing and characterizing engineered nanoparticles, resulting in several approved drugs and multiple promising candidates in clinical trials. Regulatory agencies such as the Food and Drug Administration and the European Medicines Agency released important guidance documents facilitating nanoparticle-based drug product development, particularly in the context of liposomes and lipid-based carriers. Even with the progress achieved, it is clear that many barriers must still be overcome to accelerate translation into the clinic. At the recent conference workshop "Mechanisms and Barriers in Nanomedicine" in May 2023 in Colorado, U.S.A., leading experts discussed the formulation, physiological, immunological, regulatory, clinical, and educational barriers. This position paper invites open, unrestricted, nonproprietary discussion among senior faculty, young investigators, and students to trigger ideas and concepts to move the field forward.


Asunto(s)
Nanomedicina , Humanos , Portadores de Fármacos/química , Liposomas/química , Nanopartículas/química , Estados Unidos
5.
ACS Appl Mater Interfaces ; 14(51): 56613-56622, 2022 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-36521233

RESUMEN

Novel approaches are required to address the urgent need to develop lipid-based carriers of paclitaxel (PTX) and other hydrophobic drugs for cancer chemotherapy. Carriers based on cationic liposomes (CLs) with fluid (i.e., chain-melted) membranes (e.g., EndoTAG-1) have shown promise in preclinical and late-stage clinical studies. Recent work found that the addition of a cone-shaped poly(ethylene glycol)-lipid (PEG-lipid) to PTX-loaded CLs (CLsPTX) promotes a transition to sterically stabilized, higher-curvature (smaller) nanoparticles consisting of a mixture of PEGylated CLsPTX and PTX-containing fluid lipid nanodiscs (nanodiscsPTX). These CLsPTX and nanodiscsPTX show significantly improved uptake and cytotoxicity in cultured human cancer cells at PEG coverage in the brush regime (10 mol % PEG-lipid). Here, we studied the PTX loading, in vivo circulation half-life, and biodistribution of systemically administered CLsPTX and nanodiscsPTX and assessed their ability to induce apoptosis in triple-negative breast-cancer-bearing immunocompetent mice. We focused on fluid rather than solid lipid nanodiscs because of the significantly higher solubility of PTX in fluid membranes. At 5 and 10 mol % of a PEG-lipid (PEG5K-lipid, molecular weight of PEG 5000 g/mol), the mixture of PEGylated CLsPTX and nanodiscsPTX was able to incorporate up to 2.5 mol % PTX without crystallization for at least 20 h. Remarkably, compared to preparations containing 2 and 5 mol % PEG5K-lipid (with the PEG chains in the mushroom regime), the particles at 10 mol % (with PEG chains in the brush regime) showed significantly higher blood half-life, tumor penetration, and proapoptotic activity. Our study suggests that increasing the PEG coverage of CL-based drug nanoformulations can improve their pharmacokinetics and therapeutic efficacy.


Asunto(s)
Antineoplásicos Fitogénicos , Neoplasias de la Mama , Ratones , Humanos , Animales , Femenino , Paclitaxel/química , Liposomas/química , Distribución Tisular , Caspasa 3 , Polietilenglicoles/química , Lípidos , Neoplasias de la Mama/tratamiento farmacológico , Portadores de Fármacos/química , Línea Celular Tumoral , Antineoplásicos Fitogénicos/química
6.
Int J Pharm ; 597: 120346, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33545283

RESUMEN

Breast cancer, with around 2 million new cases in 2019, is the second most common cancer worldwide and the second leading cause of cancer death among females. The aim of this work is to prepare a targeting nanoparticle through the conjugation of LinTT1 peptide, a specific molecule targeting p32 protein overexpressed by breast cancer and cancer associated cells, on liposomes' surface. This approach increases the cytotoxic effects of doxorubicin (DOX) and sorafenib (SRF) co-loaded in therapeutic liposomes on both 2D and 3D breast cancer cellular models. The liposome functionalization leads to a higher interaction with 3D breast cancer spheroids than bare ones. Moreover, interaction studies between LinTT1-functionalized liposomes and M2 primary human macrophages show an internalization of 50% of the total nanovesicles that interact with these cells, while the other 50% results only associated to cell surface. This finding suggests the possibility to use the amount of associated liposomes to enrich the hypoxic tumor area, exploiting the ability of M2 macrophages to accumulate in the central core of tumor mass. These promising results highlight the potential use of DOX and SRF co-loaded LinTT1-functionalized liposomes as nanomedicines for the treatment of breast cancer, especially in triple negative cancer cells.


Asunto(s)
Neoplasias de la Mama , Liposomas , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Doxorrubicina/uso terapéutico , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Péptidos/uso terapéutico
7.
Acta Biomater ; 133: 231-243, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33011297

RESUMEN

Nanomedicines represent innovative and promising alternative technologies to improve the therapeutic effects of different drugs for cancer ablation. Targeting M2-like tumor-associated macrophages (TAMs) has emerged as a favorable therapeutic approach to fight against cancer through the modulation of the tumor microenvironment. However, the immunomodulatory molecules used for this purpose present side effects upon systemic administration, which limits their clinical translation. Here, the biocompatible lignin polymer is used to prepare lignin nanoparticles (LNPs) that carry a dual agonist of the toll-like receptors TLR7/8 (resiquimod, R848). These LNPs are targeted to the CD206-positive M2-like TAMs using the "mUNO" peptide, in order to revert their pro-tumor phenotype into anti-tumor M1-like macrophages in the tumor microenvironment of an aggressive triple-negative in vivo model of breast cancer. Overall, we show that targeting the resiquimod (R848)-loaded LNPs to the M2-like macrophages, using very low doses of R848, induces a profound shift in the immune cells in the tumor microenvironment towards an anti-tumor immune state, by increasing the representation of M1-like macrophages, cytotoxic T cells, and activated dendritic cells. This effect consequently enhances the anticancer effect of the vinblastine (Vin) when co-administered with R848-loaded LNPs. STATEMENT OF SIGNIFICANCE: Lignin-based nanoparticles (LNPs) were successfully developed to target a potent TLR7/8 agonist (R848) of the tumor microenvironment (TME). This was achieved by targeting the mannose receptor (CD206) on the tumor supportive (M2-like) macrophages with the "mUNO" peptide, to reprogram them into an anti-tumor (M1-like) phenotype for enhanced chemotherapy. LNPs modified the biodistribution of the R848, and enhanced its accumulation and efficacy in shifting the immunological profile of the cells in the TME, which was not achieved by systemic administration of free R848. Moreover, a reduction in the tumor volumes was observed at lower equivalent doses of R848 compared with other studies. Therefore, the co-administration of R848@LNPs is a promising chemotherapeutic application in aggressive tumors, such as the triple-negative breast cancer.


Asunto(s)
Neoplasias de la Mama , Nanopartículas , Femenino , Humanos , Imidazoles , Lignina , Péptidos , Fenotipo , Distribución Tisular , Microambiente Tumoral , Macrófagos Asociados a Tumores
8.
Nanoscale ; 12(4): 2350-2358, 2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-31930241

RESUMEN

The advent of nanomedicine has recently started to innovate the treatment of cardiovascular diseases, in particular myocardial infarction. Although current approaches are very promising, there is still an urgent need for advanced targeting strategies. In this work, the exploitation of macrophage recruitment is proposed as a novel and synergistic approach to improve the addressability of the infarcted myocardium achieved by current peptide-based heart targeting strategies. For this purpose, an acetalated dextran-based nanosystem is designed and successfully functionalized with two different peptides, atrial natriuretic peptide (ANP) and linTT1, which target, respectively, cardiac cells and macrophages associated with atherosclerotic plaques. The biocompatibility of the nanocarrier is screened on both macrophage cell lines and primary macrophages, showing high safety, in particular after functionalization of the nanoparticles' surface. Furthermore, the system shows higher association versus uptake ratio towards M2-like macrophages (approximately 2-fold and 6-fold increase in murine and human primary M2-like macrophages, respectively, compared to M1-like). Overall, the results demonstrate that the nanosystem has potential to exploit the "hitchhike" effect on M2-like macrophages and potentially improve, in a dual targeting strategy, the ability of the ANP peptide to target infarcted heart.


Asunto(s)
Dextranos/química , Macrófagos/metabolismo , Infarto del Miocardio/terapia , Nanomedicina/métodos , Nanopartículas/química , Péptidos/química , Animales , Apoptosis , Factor Natriurético Atrial/química , Materiales Biocompatibles/metabolismo , Línea Celular , Humanos , Concentración de Iones de Hidrógeno , Ratones , Monocitos/metabolismo , Miocardio/metabolismo , Placa Aterosclerótica/metabolismo , Células RAW 264.7
9.
Biomaterials ; 166: 52-63, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29544111

RESUMEN

Cationic liposome-nucleic acid (CL-NA) complexes, which form spontaneously, are a highly modular gene delivery system. These complexes can be sterically stabilized via PEGylation [PEG: poly (ethylene glycol)] into nanoparticles (NPs) and targeted to specific tissues and cell types via the conjugation of an affinity ligand. However, there are currently no guidelines on how to effectively navigate the large space of compositional parameters that modulate the specific and nonspecific binding interactions of peptide-targeted NPs with cells. Such guidelines are desirable to accelerate the optimization of formulations with novel peptides. Using PEG-lipids functionalized with a library of prototypical tumor-homing peptides, we varied the peptide density and other parameters (binding motif, peptide charge, CL/DNA charge ratio) to study their effect on the binding and uptake of the corresponding NPs. We used flow cytometry to quantitatively assess binding as well as internalization of NPs by cultured cancer cells. Surprisingly, full peptide coverage resulted in less binding and internalization than intermediate coverage, with the optimum coverage varying between cell lines. In, addition, our data revealed that great care must be taken to prevent nonspecific electrostatic interactions from interfering with the desired specific binding and internalization. Importantly, such considerations must take into account the charge of the peptide ligand as well as the membrane charge density and the CL/DNA charge ratio. To test our guidelines, we evaluated the in vivo tumor selectivity of selected NP formulations in a mouse model of peritoneally disseminated human gastric cancer. Intraperitoneally administered peptide-tagged CL-DNA NPs showed tumor binding, minimal accumulation in healthy control tissues, and preferential penetration of smaller tumor nodules, a highly clinically relevant target known to drive recurrence of the peritoneal cancer.


Asunto(s)
ADN , Técnicas de Transferencia de Gen , Liposomas , Nanopartículas , Péptidos , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Cationes , ADN/química , Terapia Genética/métodos , Humanos , Lípidos/química , Liposomas/química , Nanopartículas/química , Péptidos/química
10.
Theranostics ; 7(15): 3715-3731, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29109771

RESUMEN

Fetal growth restriction (FGR) in pregnancy is commonly caused by impaired uteroplacental blood flow. Vasodilators enhance uteroplacental perfusion and fetal growth in humans and animal models; however, detrimental maternal and fetal side effects have been reported. We hypothesised that targeted uteroplacental delivery of a vasodilator would enhance drug efficacy and reduce the risks associated with drug administration in pregnancy. Phage screening identified novel peptides that selectively accumulated in the uteroplacental vasculature of pregnant mice. Following intravenous injection, the synthetic peptide CNKGLRNK selectively bound to the endothelium of the uterine spiral arteries and placental labyrinth in vivo; CNKGLRNK-decorated liposomes also selectively bound to these regions. The nitric oxide donor 2-[[4-[(nitrooxy)methyl]benzoyl]thio]-benzoic acid methyl ester (SE175) induced significant relaxation of mouse uterine arteries and human placental arteries in vitro; thus, SE175 was encapsulated into these targeted liposomes and administered to healthy pregnant C57BL/6J mice or endothelial nitric oxide synthase knockout (eNOS-/-) mice, which exhibit impaired uteroplacental blood flow and FGR. Liposomes containing SE175 (0.44mg/kg) or PBS were administered on embryonic (E) days 11.5, 13.5, 15.5 and 17.5; fetal and placental weights were recorded at term and compared to mice injected with free PBS or SE175. Targeted uteroplacental delivery of SE175 had no effect on fetal weight in C57BL/6J mice, but significantly increased fetal weight and mean spiral artery diameter, and decreased placental weight, indicative of improved placental efficiency, in eNOS-/- mice; free SE175 had no effect on fetal weight or spiral artery diameter. Targeted, but not free SE175 also significantly reduced placental expression of 4-hydroxynonenal, cyclooxygenase-1 and cyclooxygenase-2, indicating a reduction in placental oxidative stress. These data suggest that exploiting vascular targeting peptides to selectively deliver SE175 to the uteroplacental vasculature may represent a novel treatment for FGR resulting from impaired uteroplacental perfusion.


Asunto(s)
Circulación Sanguínea/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Retardo del Crecimiento Fetal/tratamiento farmacológico , Péptidos/farmacocinética , Placenta/efectos de los fármacos , Útero/efectos de los fármacos , Vasodilatadores/farmacocinética , Animales , Modelos Animales de Enfermedad , Portadores de Fármacos/administración & dosificación , Femenino , Humanos , Inyecciones Intravenosas , Liposomas/administración & dosificación , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Péptidos/administración & dosificación , Embarazo , Resultado del Tratamiento , Vasodilatadores/administración & dosificación , Vasodilatadores/farmacología
11.
J Phys Chem B ; 120(26): 6439-53, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27203598

RESUMEN

Cationic liposomes (CLs) are widely studied as carriers of DNA and short-interfering RNA for gene delivery and silencing, and related clinical trials are ongoing. Optimization of transfection efficiency (TE) requires understanding of CL-nucleic acid nanoparticle (NP) interactions with cells, NP endosomal pathways, endosomal escape, and events leading to release of active nucleic acid from the lipid carrier. Here, we studied endosomal pathways and TE of surface-functionalized CL-DNA NPs in PC-3 prostate cancer cells displaying overexpressed integrin and neuropilin-1 receptors. The NPs contained RGD-PEG-lipid or RPARPAR-PEG-lipid, targeting integrin, and neuropilin-1 receptors, respectively, or control PEG-lipid. Fluorescence colocalization using Rab11-GFP and Lysotracker enabled simultaneous colocalization of NPs with recycling endosome (Rab11) and late endosome/lysosome (Rab7/Lysotracker) pathways at increasing mole fractions of pentavalent MVL5 (+5 e) at low (10 mol %), high (50 mol %), and very high (70 mol %) membrane charge density (σM). For these cationic NPs (lipid/DNA molar charge ratio, ρchg = 5), the influence of membrane charge density on pathway selection and transfection efficiency is similar for both peptide-PEG NPs, although, quantitatively, the effect is larger for RGD-PEG compared to RPARPAR-PEG NPs. At low σM, peptide-PEG NPs show preference for the recycling endosome over the late endosome/lysosome pathway. Increases in σM, from low to high, lead to decreases in colocalization with recycling endosomes and simultaneous increases in colocalization with the late endosome/lysosome pathway. Combining colocalization and functional TE data at low and high σM shows that higher TE correlates with a larger fraction of NPs colocalized with the late endosome/lysosome pathway while lower TE correlates with a larger fraction of NPs colocalized with the Rab11 recycling pathway. The findings lead to a hypothesis that increases in σM, leading to enhanced late endosome/lysosome pathway selection and higher TE, result from increased nonspecific electrostatic attractions between NPs and endosome luminal membranes, and conversely, enhanced recycling pathway for NPs and lower TE are due to weaker attractions. Surprisingly, at very high σM, the inverse relation between the two pathways observed at low and high σM breaks down, pointing to a more complex NP pathway behavior.


Asunto(s)
ADN/administración & dosificación , Endosomas/metabolismo , Liposomas/química , Nanopartículas/administración & dosificación , Transfección , Aminas , Cationes/química , Línea Celular Tumoral , ADN/química , ADN/metabolismo , Colorantes Fluorescentes , Terapia Genética/métodos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Integrinas/metabolismo , Masculino , Potenciales de la Membrana/fisiología , Nanopartículas/química , Nanopartículas/metabolismo , Neuropilina-1/metabolismo , Transfección/métodos , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
12.
Mol Cancer Ther ; 15(4): 670-9, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26880267

RESUMEN

Peritoneal carcinomatosis is present in more than 60% of gastric cancer, 40% of ovarian cancer, and 35% of colon cancer patients. It is the second most common cause of cancer-related mortality, with a median survival of 1 to 3 months. Cytoreductive surgery combined with intraperitoneal chemotherapy is the current clinical treatment, but achieving curative drug accumulation and penetration in peritoneal carcinomatosis lesions remains an unresolved challenge. Here, we used flexible and pH-sensitive polymersomes for payload delivery to peritoneal gastric (MKN-45P) and colon (CT26) carcinoma in mice. Polymersomes were loaded with paclitaxel and in vitro drug release was studied as a function of pH and time. Paclitaxel-loaded polymersomes remained stable in aqueous solution at neutral pH for up to 4 months. In cell viability assay on cultured cancer cell lines (MKN-45P, SKOV3, CT26), paclitaxel-loaded polymersomes were more toxic than free drug or albumin-bound paclitaxel (Abraxane). Intraperitoneally administered fluorescent polymersomes accumulated in malignant lesions, and immunofluorescence revealed an intense signal inside tumors with no detectable signal in control organs. A dual targeting of tumors was observed: direct (circulation-independent) penetration, and systemic, blood vessel-associated accumulation. Finally, we evaluated preclinical antitumor efficacy of paclitaxel-polymersomes in the treatment of MKN-45P disseminated gastric carcinoma using a total dose of 7 mg/kg. Experimental therapy with paclitaxel-polymersomes improved the therapeutic index of drug over free paclitaxel and Abraxane, as evaluated by intraperitoneal tumor burden and number of metastatic nodules. Our findings underline the potential utility of the polymersome platform for delivery of drugs and imaging agents to peritoneal carcinomatosis lesions. Mol Cancer Ther; 15(4); 670-9. ©2016 AACR.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Portadores de Fármacos , Paclitaxel/administración & dosificación , Polímeros , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacocinética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Doxorrubicina/administración & dosificación , Portadores de Fármacos/química , Composición de Medicamentos , Liberación de Fármacos , Estabilidad de Medicamentos , Humanos , Inyecciones Intraperitoneales , Ratones , Paclitaxel/química , Paclitaxel/farmacocinética , Neoplasias Peritoneales/tratamiento farmacológico , Neoplasias Peritoneales/patología , Polímeros/química , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Biomaterials ; 104: 247-57, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27472162

RESUMEN

Polymersomes are versatile nanoscale vesicles that can be used for cytoplasmic delivery of payloads. Recently, we demonstrated that pH-sensitive polymersomes exhibit an intrinsic selectivity towards intraperitoneal tumor lesions. A tumor homing peptide, iRGD, harbors a cryptic C-end Rule (CendR) motif that is responsible for neuropilin-1 (NRP-1) binding and for triggering extravasation and tumor penetration of the peptide. iRGD functionalization increases tumor selectivity and therapeutic efficacy of systemic drug-loaded nanoparticles in many tumor models. Here we studied whether intraperitoneally administered paclitaxel-loaded iRGD-polymersomes show improved efficacy in the treatment of peritoneal carcinomatosis. First, we demonstrated that the pH-sensitive polymersomes functionalized with RPARPAR (a prototypic CendR peptide) or iRGD internalize in the cells that express NRP-1, and that internalized polymersomes release their cargo inside the cytosol. CendR-targeted polymersomes loaded with paclitaxel were more cytotoxic on NRP-1-positive cells than on NRP-1-negative cells. In mice bearing peritoneal tumors of gastric (MKN-45P) or colon (CT26) origin, intraperitoneally administered RPARPAR and iRGD-polymersomes showed higher tumor-selective accumulation and penetration than untargeted polymersomes. Finally, iRGD-polymersomes loaded with paclitaxel showed improved efficacy in peritoneal tumor growth inhibition and in suppression of local dissemination compared to the pristine paclitaxel-polymersomes or Abraxane. Our study demonstrates that iRGD-functionalization improves efficacy of paclitaxel-polymersomes for intraperitoneal treatment of peritoneal carcinomatosis.


Asunto(s)
Nanocápsulas/administración & dosificación , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neuropilina-1/metabolismo , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacocinética , Paclitaxel/administración & dosificación , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Línea Celular Tumoral , Sinergismo Farmacológico , Infusiones Parenterales , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanocápsulas/química , Nanoconjugados/administración & dosificación , Nanoconjugados/química , Paclitaxel/química , Polímeros/química , Resultado del Tratamiento
14.
Sci Adv ; 2(5): e1600349, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27386551

RESUMEN

The availability of therapeutics to treat pregnancy complications is severely lacking mainly because of the risk of causing harm to the fetus. As enhancement of placental growth and function can alleviate maternal symptoms and improve fetal growth in animal models, we have developed a method for targeted delivery of payloads to the placenta. We show that the tumor-homing peptide sequences CGKRK and iRGD bind selectively to the placental surface of humans and mice and do not interfere with normal development. Peptide-coated nanoparticles intravenously injected into pregnant mice accumulated within the mouse placenta, whereas control nanoparticles exhibited reduced binding and/or fetal transfer. We used targeted liposomes to efficiently deliver cargoes of carboxyfluorescein and insulin-like growth factor 2 to the mouse placenta; the latter significantly increased mean placental weight when administered to healthy animals and significantly improved fetal weight distribution in a well-characterized model of fetal growth restriction. These data provide proof of principle for targeted delivery of drugs to the placenta and provide a novel platform for the development of placenta-specific therapeutics.


Asunto(s)
Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Fragmentos de Péptidos/metabolismo , Placenta/metabolismo , Secuencia de Aminoácidos , Animales , Calreticulina/genética , Micropartículas Derivadas de Células , Femenino , Humanos , Factor II del Crecimiento Similar a la Insulina/química , Liposomas , Ratones , Nanopartículas/administración & dosificación , Nanopartículas/química , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Placenta/efectos de los fármacos , Embarazo , Unión Proteica , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética
15.
PLoS One ; 8(1): e54611, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23349939

RESUMEN

BACKGROUND: Peptide amphiphiles (PAs) are a class of amphiphilic molecules able to self-assemble into nanomaterials that have shown efficient in vivo targeted delivery. Understanding the interactions of PAs with cells and the mechanisms of their internalization and intracellular trafficking is critical in their further development for therapeutic delivery applications. METHODOLOGY/PRINCIPAL FINDINGS: PAs of a novel, cell- and tissue-penetrating peptide were synthesized possessing two different lipophilic tail architectures and their interactions with prostate cancer cells were studied in vitro. Cell uptake of peptides was greatly enhanced post-modification. Internalization occurred via lipid-raft mediated endocytosis and was common for the two analogs studied. On the contrary, we identified the non-peptidic part as the determining factor of differences between intracellular trafficking and retention of PAs. PAs composed of di-stearyl lipid tails linked through poly(ethylene glycol) to the peptide exhibited higher exocytosis rates and employed different recycling pathways compared to ones consisting of di-palmitic-coupled peptides. As a result, cell association of the former PAs decreased with time. CONCLUSIONS/SIGNIFICANCE: Control over peptide intracellular localization and retention is possible by appropriate modification with synthetic hydrophobic tails. We propose this as a strategy to design improved peptide-based delivery systems.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanoestructuras/química , Péptidos/química , Transporte de Proteínas , Línea Celular Tumoral , Exocitosis/efectos de los fármacos , Humanos , Masculino , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo , Péptidos/farmacología , Polietilenglicoles/química , Neoplasias de la Próstata/tratamiento farmacológico , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología
16.
Science ; 328(5981): 1031-5, 2010 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-20378772

RESUMEN

Poor penetration of anticancer drugs into tumors can be an important factor limiting their efficacy. We studied mouse tumor models to show that a previously characterized tumor-penetrating peptide, iRGD, increased vascular and tissue permeability in a tumor-specific and neuropilin-1-dependent manner, allowing coadministered drugs to penetrate into extravascular tumor tissue. Importantly, this effect did not require the drugs to be chemically conjugated to the peptide. Systemic injection with iRGD improved the therapeutic index of drugs of various compositions, including a small molecule (doxorubicin), nanoparticles (nab-paclitaxel and doxorubicin liposomes), and a monoclonal antibody (trastuzumab). Thus, coadministration of iRGD may be a valuable way to enhance the efficacy of anticancer drugs while reducing their side effects, a primary goal of cancer therapy research.


Asunto(s)
Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias/tratamiento farmacológico , Oligopéptidos/administración & dosificación , Paclitaxel Unido a Albúmina , Albúminas/administración & dosificación , Albúminas/farmacocinética , Albúminas/uso terapéutico , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Permeabilidad Capilar/efectos de los fármacos , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapéutico , Humanos , Liposomas , Ratones , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neuropilina-1/metabolismo , Oligopéptidos/metabolismo , Oligopéptidos/farmacocinética , Oligopéptidos/farmacología , Paclitaxel/administración & dosificación , Paclitaxel/farmacocinética , Paclitaxel/uso terapéutico , Permeabilidad , Trastuzumab , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA