Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Colloids Surf B Biointerfaces ; 167: 474-482, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29723819

ABSTRACT

One of the major challenges in the administration of therapeutic proteins involves delivery limitations. Liposomes are well-known drug delivery systems (DDS) that have been used to overcome this drawback; nevertheless, low protein entrapment efficiency (EE) still limits their wide biomedical application on a commercial scale. In the present work, different methods for protein entrapment into liposomes were tested in order to obtain tailored DDS platforms for multiple biomedical applications. The protein used as model was the Black-eyed pea Trypsin and Chymotrypsin Inhibitor (BTCI), a member of the Bowman-Birk protease inhibitor family (BBIs), which has been largely explored for its potential application in many biomedical therapies. We optimized reverse-phase evaporation (REV) and freeze/thaw (F/T) entrapment methods, using a cationic lipid matrix to entrap expressive amounts of BTCI (∼100 µM) in stable liposomes without affecting its protease inhibition activity. The influence of various parameters (e.g. entrapment method, liposome composition, buffer type) on particle size, charge, polydispersity, and EE of liposomes was investigated to provide an insight on how to control such parameters in view of obtaining a high entrapment yield. In addition, BTCI liposome platforms obtained herein showed to be versatile vesicles, allowing surface modification with moieties/polymers of interest (e.g. PEG, transferrin). The aforementioned results are relevant to focusing on the entrapment of other promising BBIs or protein agents sharing similar structural features. These findings encourage future studies to investigate the advantages of using the liposome platforms presented herein to broaden the use of this type of DDS for BBI biomedical applications.


Subject(s)
Drug Delivery Systems/methods , Liposomes/chemistry , Vigna/metabolism , Biocatalysis/drug effects , Chymotrypsin/metabolism , Particle Size , Plant Proteins/administration & dosage , Plant Proteins/chemistry , Polyethylene Glycols/chemistry , Surface Properties , Transferrin/chemistry , Trypsin/metabolism
2.
Acta Biomater ; 55: 443-454, 2017 06.
Article in English | MEDLINE | ID: mdl-28400314

ABSTRACT

A novel drug delivery system for doxorubicin (DOX), based on organic-inorganic composites was developed. DOX was incorporated in micelles (M-DOX) of polyethylene glycol-phosphatidylethanolamine (PEG-PE) which in turn were adsorbed by the clay, montmorillonite (MMT). The nano-structures of the PEG-PE/MMT composites of LOW and HIGH polymer loadings were characterized by XRD, TGA, FTIR, size (DLS) and zeta measurements. These measurements suggest that for the LOW composite a single layer of polymer intercalates in the clay platelets and the polymer only partially covers the external surface, while for the HIGH composite two layers of polymer intercalate and a bilayer may form on the external surface. These nanostructures have a direct effect on formulation stability and on the rate of DOX release. The release rate was reversely correlated with the degree of DOX interaction with the clay and followed the sequence: M-DOX>HIGH formulation>LOW formulation>DOX/MMT. Despite the slower release from the HIGH formulation, its cytotoxicity effect on sensitive cells was as high as the "free" DOX. Surprisingly, the LOW formulation, with the slowest release, demonstrated the highest cytotoxicity in the case of Adriamycin (ADR) resistant cells. Confocal microscopy images and association tests provided an insight into the contribution of formulation-cell interactions vs. the contribution of DOX release rate. Internalization of the formulations was suggested as a mechanism that increases DOX efficiency, particularly in the ADR resistant cell line. The employment of organic-inorganic hybrid materials as drug delivery systems, has not reached its full potential, however, its functionality as an efficient tunable release system was demonstrated. STATEMENT OF SIGNIFICANCE: DOX PEG-PE/clay formulations were design as an efficient drug delivery system. The main aim was to develop PEG-PE/clay formulations of different structures based on various PEG-PE/clay ratios in order to achieve tunable release rates, to control the external surface characteristics and formulation stability. The formulations showed significantly higher toxicity in comparison to "free" DOX, explained by formulation internalization. For each cell line tested, sensitive and ADR resistant, a different formulation structure was found most efficient. The potential of PEG-PE/clay-DOX formulations to improve DOX administration efficacy was demonstrated and should be further explored and implemented for other cancer drugs and cells.


Subject(s)
Aluminum Silicates , Cytotoxins , Doxorubicin , Drug Carriers , Neoplasms/drug therapy , Polyethylene Glycols , Aluminum Silicates/chemistry , Aluminum Silicates/pharmacokinetics , Aluminum Silicates/pharmacology , Bentonite/chemistry , Bentonite/pharmacokinetics , Bentonite/pharmacology , Cell Line, Tumor , Clay , Cytotoxins/chemistry , Cytotoxins/pharmacokinetics , Cytotoxins/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Humans , Neoplasms/metabolism , Neoplasms/pathology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/pharmacology
3.
Acta Biomater ; 35: 248-59, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26884277

ABSTRACT

Future improvements of non-viral vectors for siRNA delivery require better understanding of intracellular processing and vector interactions with target cells. Here, we have compared the siRNA delivery properties of a lipid derivative of bPEI 1.8kDa (DOPE-PEI) with branched polyethyleneimine (bPEI) with average molecular weights of 1.8kDa (bPEI 1.8kDa) and 25kDa (bPEI 25kDa). We find mechanistic differences between the DOPE-PEI conjugate and bPEI regarding siRNA condensation and intracellular processing. bPEI 1.8kDa and bPEI 25kDa have similar properties with respect to condensation capability, but are very different regarding siRNA decondensation, cellular internalization and induction of reporter gene knockdown. Lipid conjugation of bPEI 1.8kDa improves the siRNA delivery properties, but with markedly different formulation requirements and mechanisms of action compared to conventional PEIs. Interestingly, strong knockdown using bPEI 25kDa is dependent on the presence of a free vector fraction which does not increase siRNA uptake. Finally, we have investigated the effect on lysosomal pH induced by these vectors to elucidate the differences in the proton sponge effect between lipid conjugated PEI and conventional PEI: Neither DOPE-PEI nor bPEI 25kDa affected lysosomal pH as a function of time, underlining that the possible proton sponge effect is not associated with changes in lysosomal pH. STATEMENT OF SIGNIFICANCE: Gene silencing therapy has the potential to treat diseases which are beyond the reach of current small molecule-based medicines. However, delivery of the small interfering RNAs (siRNAs) remains a bottleneck to clinical implementation, and the development of safe and efficient delivery systems would be one of the most important achievements in medicine today. A major reason for the lack of progress is insufficient understanding of cell-polyplex interaction. We investigate siRNA delivery using polyethyleneimine (PEI) based vectors and examine how crucial formulation parameters determine the challenges associated with PEI as a delivery vector. We further evaluate how lipid conjugation of PEI influences formulation, cytotoxicity and polymer interaction with cells and cargo as well as the proton sponge capabilities of the vectors.


Subject(s)
Gene Knockdown Techniques , Polyamines/metabolism , RNA, Small Interfering/metabolism , Cell Death/drug effects , Cell Line, Tumor , Endocytosis/drug effects , Genetic Vectors/metabolism , Heparin/pharmacology , Humans , Hydrogen-Ion Concentration , Luciferases/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Particle Size , Polyelectrolytes , Static Electricity
4.
Pharm Res ; 32(8): 2753-63, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25732960

ABSTRACT

PURPOSE: Platinum-based therapies are the first line treatments for most types of cancer including ovarian cancer. However, their use is associated with dose-limiting toxicities and resistance. We report initial translational studies of a theranostic nanoemulsion loaded with a cisplatin derivative, myrisplatin and pro-apoptotic agent, C6-ceramide. METHODS: The surface of the nanoemulsion is annotated with an endothelial growth factor receptor (EGFR) binding peptide to improve targeting ability and gadolinium to provide diagnostic capability for image-guided therapy of EGFR overexpressing ovarian cancers. A high shear microfludization process was employed to produce the formulation with particle size below 150 nm. RESULTS: Pharmacokinetic study showed a prolonged blood platinum and gadolinium levels with nanoemulsions in nu/nu mice. The theranostic nanoemulsions also exhibited less toxicity and enhanced the survival time of mice as compared to an equivalent cisplatin treatment. CONCLUSIONS: Magnetic resonance imaging (MRI) studies indicate the theranostic nanoemulsions were effective contrast agents and could be used to track accumulation in a tumor. The MRI study additionally indicate that significantly more EGFR-targeted theranostic nanoemulsion accumulated in a tumor than non-targeted nanoemulsuion providing the feasibility of using a targeted theranostic agent in conjunction with MRI to image disease loci and quantify the disease progression.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Ceramides/administration & dosage , Ceramides/therapeutic use , ErbB Receptors/drug effects , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/therapeutic use , Ovarian Neoplasms/drug therapy , Theranostic Nanomedicine/methods , Animals , Antineoplastic Agents/pharmacokinetics , Blood Platelets/metabolism , Ceramides/pharmacokinetics , Drug Delivery Systems , Female , Gadolinium/metabolism , Mice , Microfluidics , Organoplatinum Compounds/pharmacokinetics , Particle Size , Survival Analysis , Tissue Distribution
5.
J Vasc Interv Radiol ; 25(12): 1972-82, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25439675

ABSTRACT

PURPOSE: To characterize upregulation of hypoxia-inducible factor (HIF)-1α after radiofrequency (RF) ablation and the influence of an adjuvant HIF-1α inhibitor (bortezomib) and nanodrugs on modulating RF ablation-upregulated hypoxic pathways. MATERIALS AND METHODS: Fisher 344 rats (n = 68) were used. First, RF ablation-induced periablational HIF-1α expression was evaluated in normal liver or subcutaneous R3230 tumors (14-16 mm). Next, the effect of varying RF ablation thermal dose (varying tip temperature 50°C-90°C for 2-20 minutes) on HIF-1α expression was studied in R3230 tumors. Third, RF ablation was performed in R3230 tumors without or with an adjuvant HIF-1α inhibitor, bortezomib (single intraperitoneal dose 0.1 mg/kg). Finally, the combination RF ablation and intravenous liposomal chemotherapeutics with known increases in periablational cellular cytotoxicity (doxorubicin, paclitaxel, and quercetin) was assessed for effect on periablational HIF-1α. Outcome measures included immunohistochemistry of HIF-1α and heat shock protein 70 (marker of nonlethal thermal injury). RESULTS: RF ablation increased periablational HIF-1α in both normal liver and R3230 tumor, peaking at 24-72 hours. Tumor RF ablation had similar HIF-1α rim thickness but significantly greater percent cell positivity compared with hepatic RF ablation (P < .001). HIF-1α after ablation was the same regardless of thermal dose. Bortezomib suppressed HIF-1α (rim thickness, 68.7 µm ± 21.5 vs 210.3 µm ± 85.1 for RF ablation alone; P < .02) and increased ablation size (11.0 mm ± 1.5 vs 7.7 mm ± 0.6 for RF ablation alone; P < .002). Finally, all three nanodrugs suppressed RF ablation-induced HIF-1α (ie, rim thickness and cell positivity; P < .02 for all comparisons), with liposomal doxorubicin suppressing HIF-1α the most (P < .03). CONCLUSIONS: RF ablation upregulates HIF-1α in normal liver and tumor in a temperature-independent manner. This progrowth, hypoxia pathway can be successfully suppressed with an adjuvant HIF-1α-specific inhibitor, bortezomib, or non-HIF-1α-specific liposomal chemotherapy.


Subject(s)
Boronic Acids/pharmacology , Breast Neoplasms/surgery , Catheter Ablation/methods , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Liposomes/pharmacology , Pyrazines/pharmacology , Up-Regulation/drug effects , Animals , Antineoplastic Agents/pharmacology , Bortezomib , Chemotherapy, Adjuvant , Disease Models, Animal , Female , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver/drug effects , Liver/surgery , Rats , Rats, Inbred F344
6.
PLoS One ; 9(8): e102727, 2014.
Article in English | MEDLINE | ID: mdl-25133740

ABSTRACT

PURPOSE: To determine the effect of different drug-loaded nanocarriers (micelles and liposomes) on delivery and treatment efficacy for radiofrequency ablation (RFA) combined with nanodrugs. MATERIALS/METHODS: Fischer 344 rats were used (n = 196). First, single subcutaneous R3230 tumors or normal liver underwent RFA followed by immediate administration of i.v. fluorescent beads (20, 100, and 500 nm), with fluorescent intensity measured at 4-24 hr. Next, to study carrier type on drug efficiency, RFA was combined with micellar (20 nm) or liposomal (100 nm) preparations of doxorubicin (Dox; targeting HIF-1α) or quercetin (Qu; targeting HSP70). Animals received RFA alone, RFA with Lipo-Dox or Mic-Dox (1 mg i.v., 15 min post-RFA), and RFA with Lipo-Qu or Mic-Qu given 24 hr pre- or 15 min post-RFA (0.3 mg i.v.). Tumor coagulation and HIF-1α or HSP70 expression were assessed 24 hr post-RFA. Third, the effect of RFA combined with i.v. Lipo-Dox, Mic-Dox, Lipo-Qu, or Mic-Qu (15 min post-RFA) compared to RFA alone on tumor growth and animal endpoint survival was evaluated. Finally, drug uptake was compared between RFA/Lipo-Dox and RFA/Mic-Dox at 4-72 hr. RESULTS: Smaller 20 nm beads had greater deposition and deeper tissue penetration in both tumor (100 nm/500 nm) and liver (100 nm) (p<0.05). Mic-Dox and Mic-Qu suppressed periablational HIF-1α or HSP70 rim thickness more than liposomal preparations (p<0.05). RFA/Mic-Dox had greater early (4 hr) intratumoral doxorubicin, but RFA/Lipo-Dox had progressively higher intratumoral doxorubicin at 24-72 hr post-RFA (p<0.04). No difference in tumor growth and survival was seen between RFA/Lipo-Qu and RFA/Mic-Qu. Yet, RFA/Lipo-Dox led to greater animal endpoint survival compared to RFA/Mic-Dox (p<0.03). CONCLUSION: With RF ablation, smaller particle micelles have superior penetration and more effective local molecular modulation. However, larger long-circulating liposomal carriers can result in greater intratumoral drug accumulation over time and reduced tumor growth. Accordingly, different carriers provide specific advantages, which should be considered when formulating optimal combination therapies.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Catheter Ablation , Doxorubicin/administration & dosage , Animals , Chemoradiotherapy , Female , Liposomes , Micelles , Nanoparticles , Particle Size , Quercetin/administration & dosage , Rats, Inbred F344 , Xenograft Model Antitumor Assays
7.
Nat Med ; 20(7): 778-784, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24880615

ABSTRACT

Chemoradiation-resistant cancers limit treatment efficacy and safety. We show here the cancer cell-specific, on-demand intracellular amplification of chemotherapy and chemoradiation therapy via gold nanoparticle- and laser pulse-induced mechanical intracellular impact. Cancer aggressiveness promotes the clustering of drug nanocarriers and gold nanoparticles in cancer cells. This cluster, upon exposure to a laser pulse, generates a plasmonic nanobubble, the mechanical explosion that destroys the host cancer cell or ejects the drug into its cytoplasm by disrupting the liposome and endosome. The same cluster locally amplifies external X-rays. Intracellular synergy of the mechanical impact of plasmonic nanobubble, ejected drug and amplified X-rays improves the efficacy of standard chemoradiation in resistant and aggressive head and neck cancer by 100-fold in vitro and 17-fold in vivo, reduces the effective entry doses of drugs and X-rays to 2-6% of their clinical doses and efficiently spares normal cells. The developed quadrapeutics technology combines four clinically validated components and transforms a standard macrotherapy into an intracellular on-demand theranostic microtreatment with radically amplified therapeutic efficacy and specificity.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Nanostructures , Animals , Combined Modality Therapy , Drug Resistance, Neoplasm , Humans , Mice , Treatment Outcome
8.
Pharm Res ; 31(8): 1882-92, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24522814

ABSTRACT

PURPOSE: To develop a nanostructured lipid carrier (NLC) co-loaded with doxorubicin and docosahexaenoic acid (DHA) and to evaluate its potential to overcome drug resistance and to increase antitumoral effect in MCF-7/Adr cancer cell line. METHODS: The NLC was prepared by a hot homogenization method and characterized for size, zeta potential, entrapment efficiency (EE) and drug loading (DL). Drug release was evaluated by dialysis in complete DMEM, and NLC aggregation was assayed in the presence of serum. The cytotoxicity of formulations, doxorubicin uptake or penetration were evaluated in MCF-7 and MCF-7/Adr as monolayer or spheroid models. RESULTS: The formulation had a size of about 80 nm, negative zeta potential, EE of 99%, DL of 31 mg/g, a controlled drug release in DMEM and no particles aggregation in presence of serum. The NLC loaded with doxorubicin and DHA showed the same activity as free drugs against MCF-7 but a stronger activity against MCF-7/Adr cells. In monolayer model, the doxorubicin uptake as free and encapsulated form was similar in MCF-7 but higher for the encapsulated drug in MCF-7/Adr, suggesting a bypassing of P-glycoprotein bomb efflux. For spheroids, the NLC loaded with doxorubicin and DHA showed a prominent cytotoxicity and a greater penetration of doxorubicin. CONCLUSIONS: These findings suggest that the co-encapsulation of doxorubicin and DHA in NLC enhances the cytotoxicity and overcomes the doxorubicin resistance in MCF-7/Adr.


Subject(s)
Docosahexaenoic Acids/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Drug Resistance, Neoplasm/drug effects , Nanostructures/administration & dosage , Cell Survival/drug effects , Cell Survival/physiology , Chemistry, Pharmaceutical , Docosahexaenoic Acids/chemistry , Dose-Response Relationship, Drug , Doxorubicin/chemistry , Drug Carriers/chemistry , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/physiology , Humans , MCF-7 Cells , Nanostructures/chemistry
9.
Mol Pharm ; 11(2): 375-81, 2014 Feb 03.
Article in English | MEDLINE | ID: mdl-24325630

ABSTRACT

In this study, transferrin (Tf)-modified poly(ethylene glycol)-phosphatidylethanolamine (mPEG-PE) micelles loaded with the poorly water-soluble drug, R547 (a potent and selective ATP-competitive cyclin-dependent kinase (CDK) inhibitor), were prepared and evaluated for their targeting efficiency and cytotoxicity in vitro and in vivo to A2780 ovarian carcinoma cells, which overexpress transferrin receptors (TfR). At 10 mM lipid concentration, both Tf-modified and plain micelles solubilized 800 µg of R547. Tf-modified micelles showed enhanced interaction with A2780 ovarian carcinoma cells in vitro. The involvement of TfR in endocytosis of Tf-modified micelles was confirmed by colocalization studies of micelle-treated cells with the endosomal marker Tf-Alexa488. We confirmed endocytosis of micelles in an intact form with micelles loaded with a fluorescent dye and additionally labeled with fluorescent lipid. The in vitro cytotoxicity and in vivo tumor growth inhibition studies in A2780-tumor bearing mice confirmed the enhanced efficacy of Tf-modified R547-loaded micelles compared to free drug solution and to nonmodified micelles. The results of this study demonstrate the potential application of Tf-conjugated polymeric micelles in the treatment of tumors overexpressing TfR.


Subject(s)
Drug Delivery Systems , Micelles , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Pyrimidines/chemistry , Pyrimidines/pharmacology , Transferrin/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Flow Cytometry , Humans , Mice , Microscopy, Confocal , Solubility , Water/chemistry
10.
Nanomedicine ; 10(2): 411-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23928214

ABSTRACT

The clinical application of gene silencing mediated by small interfering RNA (siRNA) has been limited by the lack of efficient and safe carriers. Phospholipid modification of low molecular weight polyethylenimine (PEI 1.8 kDa) dramatically increased its gene down-regulation capacity while keeping cytotoxicity levels low. The silencing efficacy was highly dependent on the nature of the lipid grafted to PEI and the polymer/siRNA ratio employed. Phosphoethanolamine (DOPE and DPPE) and phosphocholine (PC) conjugation did not change the physicochemical properties and siRNA binding capacity of PEI complexes but had a large impact on their transfection and ability to down-regulate Green Fluorescent Protein (GFP) expression (60%, 30% and 5% decrease of GFP expression respectively). We found that the micelle-forming structure of DOPE and DPPE-PEI dramatically changed PEI's interaction with cell membranes and played a key role in promoting PEI 1.8 kDa transfection, completely ineffective in the absence of the lipid modification. FROM THE CLINICAL EDITOR: While siRNA-based gene silencing methods could have numerous clinical applications, efficient delivery remains a major challenge. This team reports that DOPE-PEI and DPPE-PEI based micelle-forming nanostructures may be able to provide an efficient vector for siRNA transfection.


Subject(s)
Lipids/chemistry , Nanoparticles/chemistry , Phospholipids/chemistry , Polyethyleneimine/chemistry , RNA Interference , Animals , Cell Line , Cell Membrane/metabolism , Drug Carriers/chemistry , Ethanolamines/chemistry , Green Fluorescent Proteins/metabolism , Humans , Inhibitory Concentration 50 , Mice , Micelles , Molecular Weight , Nanomedicine , Phosphorylcholine/chemistry , RNA, Small Interfering/chemistry
11.
J Vasc Interv Radiol ; 24(7): 1021-33, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23664809

ABSTRACT

PURPOSE: To evaluate the effects of radiofrequency (RF) ablation without and with adjuvant intravenous (IV) liposomal doxorubicin (Doxil) on microvessel morphology and patency and intratumoral drug delivery and retention. MATERIALS AND METHODS: There were 133 tumors/animals used in this experiment. First, single subcutaneous tumors (R3230 in Fischer rats and 786-0 in nude mice) were randomly assigned to receive RF ablation alone or no treatment and sacrificed 0-72 hours after treatment. Next, combined RF ablation and liposomal doxorubicin (1 mg given 15 min after RF ablation) was studied in R3230 tumors at 0-72 hours after treatment. Histopathologic assessment, including immunohistochemical staining for cleaved caspase-3, heat-shock protein 70, and CD34, was performed to assess morphologic vessel appearance, vessel diameter, and microvascular density. Subsequently, tumors were randomly assigned to receive RF ablation alone, RF ablation and liposomal doxorubicin, or no treatment (control tumors), followed by IV fluorescent-labeled liposomes (a surrogate marker) given 0-24 hours after RF ablation to permit qualitative assessment. RESULTS: RF ablation alone resulted in enlarged and dysmorphic vessels from 0-4 hours, peaking at 12-24 hours after RF ablation, occurring preferentially closer to the electrode. The addition of doxorubicin resulted in earlier vessel contraction (mean vessel area, 47,539 µm(2)±9,544 vs 1,854 µm(2)±458 for RF ablation alone at 15 min; P<.05). Combined RF ablation and liposomal doxorubicin produced similar fluorescence 1 hour after treatment (40.88 AU/µm(2)±33.53 vs 22.1 AU/µm(2)±13.19; P = .14) but significantly less fluorescence at 4 hours (24.3 AU/µm(2)±3.65 vs 2.8 AU/µm(2)±3.14; P<.002) compared with RF ablation alone denoting earlier reduction in microvascular patency. CONCLUSIONS: RF ablation induces morphologic changes to vessels within the ablation zone lasting 12-24 hours after treatment. The addition of liposomal doxorubicin causes early vessel contraction and a reduction in periablational microvascular patency. Such changes would likely need to be considered when determining optimal drug administration and imaging paradigms.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Catheter Ablation , Doxorubicin/analogs & derivatives , Kidney Neoplasms/drug therapy , Kidney Neoplasms/surgery , Microvessels/drug effects , Administration, Intravenous , Animals , Antibiotics, Antineoplastic/administration & dosage , Biomarkers, Tumor/metabolism , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Catheter Ablation/adverse effects , Cell Line, Tumor , Chemotherapy, Adjuvant , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Female , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Nude , Microvessels/metabolism , Microvessels/pathology , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacology , Rats , Rats, Inbred F344 , Time Factors , Xenograft Model Antitumor Assays
12.
J Drug Target ; 21(7): 630-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23594094

ABSTRACT

We prepared and evaluated transferrin (Tf) and monoclonal antibody (mAb) 2C5-modified dual ligand-targeted poly(ethylene glycol)-phosphatidylethanolamine micelles loaded with a poorly soluble drug, R547 (a selective adenosine triphosphate-competitive cyclin-dependent kinase inhibitor) for enhancement of targeting efficiency and cytotoxicity in vitro and in vivo to A2780 ovarian carcinoma compared to single ligand-targeted micelles. Micellar solubilization significantly improved the solubility of R547 from 1 to 800 µg/mL. The size of modified and non-modified micelles was 13-16 nm. Flow cytometry indicated significantly enhanced cellular association of dual ligand-targeted micelles compared to single ligand-targeted micelles. Confocal microscopy confirmed the Tf receptor-mediated endocytosis of rhodamine-labeled Tf-modified micelles after staining the micelle-treated cells with the endosomal marker Tf-Alexa488. The optimized dual-targeted micelles enhanced cytotoxicity in vitro against A2780 ovarian cancer cells compared to plain and single ligand-targeted micelles. Interestingly, in vivo anti-tumor efficacy was more pronounced for the preparation with a single-targeting ligand (Tf). The specific combination Tf and mAb 2C5 did not yield the expected increase in efficacy as was observed in vitro. This observation suggests that the relationships between targeting ligands in vivo could be more complex than in simplified in vitro systems, and the results of the optimization process should always be verified in vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Micelles , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Cells, Cultured , Drug Screening Assays, Antitumor , Flow Cytometry , Humans , Ligands , Microscopy, Confocal , Solubility
13.
Adv Drug Deliv Rev ; 65(1): 24-35, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22964425

ABSTRACT

Significant progress has been made recently in the area of immunoconjugated drugs and drug delivery systems (DDS). The immuno-modification of either the drug or DDS has proven to be a very promising approach that has significantly improved the targeted accumulation in pathological sites while decreasing its undesirable side effects in healthy tissues. The arrangement for both prolonged life in the circulation and specific target recognition represents another potent strategy in the development of immuno-targeted systems. The longevity of immuno-targeted DDS such as immunoliposomes and immunomicelles improves their targetability even in the presence of the additional passive accumulation in areas with a compromised vasculature. The added use of the immuno-targeted systems takes advantage of the specific microenvironment of pathological sites including lowered pH, increased temperature, and variation in the enzymatic activity. "Smart" stimulus-responsive systems combine different valuable functionalities including PEG-protection, targeting antibody, cell-penetration, and stimulus-sensitive functions. In this review we examined the evolution, current status and future directions in the area of therapeutical immunoconjugates and long-circulating immuno-targeted DDS.


Subject(s)
Drug Delivery Systems/methods , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems/trends , Forecasting , Humans , Immunoconjugates/pharmacokinetics , Liposomes/administration & dosage , Liposomes/chemistry , Liposomes/pharmacokinetics , Micelles , Pharmacokinetics
14.
J Biophotonics ; 6(5): 425-34, 2013 May.
Article in English | MEDLINE | ID: mdl-22903924

ABSTRACT

In vivo photoacoustic (PA) and fluorescence flow cytometry were previously applied separately using pulsed and continuous wave lasers respectively, and positive contrast detection mode only. This paper introduces a real-time integration of both techniques with positive and negative contrast modes using only pulsed lasers. Various applications of this new tool are summarized, including detection of liposomes loaded with Alexa-660 dye, red blood cells labeled with Indocyanine Green, B16F10 melanoma cells co-expressing melanin and green fluorescent protein (GFP), C8161-GFP melanoma cells targeted by magnetic nanoparticles, MTLn3 adenocarcinoma cells expressing novel near-infrared iRFP protein, and quantum dot-carbon nanotube conjugates. Negative contrast flow cytometry provided label-free detection of low absorbing or weakly fluorescent cells in blood absorption and autofluorescence background, respectively. The use of pulsed laser for time-resolved discrimination of objects with long fluorescence lifetime (e.g., quantum dots) from shorter autofluorescence background (e.g., blood plasma) is also highlighted in this paper. The supplementary nature of PA and fluorescence detection increased the versatility of the integrated method for simultaneous detection of probes and cells having various absorbing and fluorescent properties, and provided verification of PA data using a more established fluorescence based technique.


Subject(s)
Flow Cytometry/methods , Fluorescence , Neoplastic Cells, Circulating/pathology , Optical Phenomena , Photoacoustic Techniques/methods , Animals , Cell Line, Tumor , Humans , Lasers , Mice , Nanoparticles/metabolism
15.
Adv Drug Deliv Rev ; 64(13): 1436-46, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22917778

ABSTRACT

Personalized medicine, which ultimately seeks to afford tailored therapeutic regimens for individual patients, is quickly emerging as a new paradigm in the diagnosis and treatment of diseases. The idea of casting aside generic treatments in favor of patient-centric therapies has become feasible owing to advances in nanotechnology and drug delivery coupled with an enhanced knowledge of genomics and an understanding of disease at the molecular level. This review highlights polymeric immunomicelles as a class of nanocarriers that have the potential to combine diagnosis, targeted drug therapy, as well as imaging and monitoring of therapeutic response, to render a personalized approach to the management of disease. Smart multi-functional immunomicelles, as the next generation of nanocarriers, are poised for facilitating personalized cancer treatment. This review provides an assessment of immunomicelles as tools for advancing personalized therapy of diseases, with cancer being the major focus.


Subject(s)
Drug Carriers/administration & dosage , Micelles , Nanomedicine , Precision Medicine , Animals , Antineoplastic Agents/administration & dosage , Humans , Neoplasms/drug therapy , Plaque, Atherosclerotic/diagnosis , Polymers/administration & dosage , RNA, Small Interfering/administration & dosage
16.
AAPS J ; 14(2): 303-15, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22415612

ABSTRACT

Liposomes, phospholipid vesicles with a bilayered membrane structure, have been widely used as pharmaceutical carriers for drugs and genes, in particular for treatment of cancer. To enhance the efficacy of the liposomal drugs, drug-loaded liposomes are targeted to the tumors by means of passive (enhanced permeability and retention mediated) targeting, based on the longevity of liposomes in blood and its accumulation in pathological sites with compromised vasculature, and active targeting, based on the attachment of specific ligands to the liposomal surface to bind certain antigens on the target cells. Antibody-targeted liposomes loaded with anticancer drugs demonstrate high potential for clinical applications. This review highlights evolution of liposomes for both passive and active targeting and challenges in development of targeted liposomal therapeutics specifically antibody-targeted liposomes.


Subject(s)
Drug Delivery Systems/trends , Drug Discovery/trends , Liposomes/administration & dosage , Pharmaceutical Preparations/administration & dosage , Animals , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems/methods , Drug Discovery/methods , Humans , Liposomes/chemistry , Pharmaceutical Preparations/chemistry
17.
Bioconjug Chem ; 23(4): 785-95, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22404783

ABSTRACT

As part of our program to develop breast cancer specific therapeutic agents, we have synthesized a conjugate agent that is a conjugate of the steroidal anti-estrogen and the potent cytotoxin doxorubicin. In this effort, we employed a modular assembly approach to prepare a novel 11ß-substituted steroidal anti-estrogen functionalized with an azido-tetraethylene glycol moiety, which could be coupled to a complementary doxorubicin benzoyl hydrazone functionalized with a propargyl tetraethylene glycol moiety. Huisgen [3 + 2] cycloaddition chemistry gave the final hybrid that was evaluated for selective uptake and cytotoxicity in ER(+)-MCF-7 and ER(-)-MDA-MB-231 breast cancer cell lines. The results demonstrated that the presence of the anti-estrogenic component in the hybrid compound was critical for selectivity and cytotoxicity in ER(+)-MCF-7 human breast cancer cells as the hybrid was ~70-fold more potent than doxorubicin in inhibition of cell proliferation and promoting cell death.


Subject(s)
Breast Neoplasms/metabolism , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Drug Design , Estrogen Antagonists/chemistry , Molecular Targeted Therapy , Receptors, Estrogen/metabolism , Steroids/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Doxorubicin/chemical synthesis , Doxorubicin/pharmacology , Humans , Inhibitory Concentration 50
18.
Biomaterials ; 33(15): 3942-51, 2012 May.
Article in English | MEDLINE | ID: mdl-22365809

ABSTRACT

A low molecular weight polyethyleneimine (PEI 1.8 kDa) was modified with dioleoylphosphatidylethanolamine (PE) to form the PEI-PE conjugate investigated as a transfection vector. The optimized PEI-PE/pDNA complexes at an N/P ratio of 16 had a particle size of 225 nm, a surface charge of +31 mV, and protected the pDNA from the action of DNase I. The PEI-PE conjugate had a critical micelle concentration (CMC) of about 34 µg/ml and exhibited no toxicity compared to a high molecular weight PEI (PEI 25 kDa) as tested with B16-F10 melanoma cells. The B16-F10 cells transfected with PEI-PE/pEGFP complexes showed protein expression levels higher than with PEI-1.8 or PEI-25 vectors. Complexes prepared with YOYO 1-labeled pEGFP confirmed the enhanced delivery of the plasmid with PEI-PE compared to PEI-1.8 and PEI-25. The PEI-PE/pDNA complexes were also mixed with various amounts of micelle-forming material, polyethylene glycol (PEG)-PE to improve biocompatibility. The resulting particles exhibited a neutral surface charge, resistance to salt-induced aggregation, and good transfection activity in the presence of serum in complete media. The use of the low-pH-degradable PEG-hydrazone-PE produced particles with transfection activity sensitive to changes in pH consistent with the relatively acidic tumor environment.


Subject(s)
Gene Transfer Techniques , Lipids/chemistry , Micelles , Phosphatidylethanolamines/chemistry , Polyethyleneimine/chemistry , Animals , Cell Death , Chromatography, High Pressure Liquid , DNA/metabolism , Electrophoresis, Agar Gel , Hydrogen-Ion Concentration , Melanoma, Experimental , Mice , Microscopy, Confocal , Particle Size , Phosphatidylethanolamines/chemical synthesis , Polyethylene Glycols/chemistry , Polyethyleneimine/chemical synthesis , Static Electricity , Transfection
19.
J Control Release ; 160(2): 239-44, 2012 Jun 10.
Article in English | MEDLINE | ID: mdl-22230341

ABSTRACT

BACKGROUND: To investigate the effect of IV liposomal quercetin (a known down-regulator of heat shock proteins) alone and with liposomal doxorubicin on tumor growth and end-point survival when combined with radiofrequency (RF) tumor ablation in a rat tumor model. METHODS: Solitary subcutaneous R3230 mammary adenocarcinoma tumors (1.3-1.5 cm) were implanted in 48 female Fischer rats. Initially, 32 tumors (n=8, each group) were randomized into four experimental groups: (a) conventional monopolar RF alone (70°C for 5 min), (b) IV liposomal quercetin alone (1 mg/kg), (c) IV liposomal quercetin followed 24hr later with RF, and (d) no treatment. Next, 16 additional tumors were randomized into two groups (n=8, each) that received a combined RF and liposomal doxorubicin (15 min post-RF, 8 mg/kg) either with or without liposomal quercetin. Kaplan-Meier survival analysis was performed using a tumor diameter of 3.0 cm as the defined survival endpoint. RESULTS: Differences in endpoint survival and tumor doubling time among the groups were highly significant (P<0.001). Endpoint survivals were 12.5±2.2 days for the control group, 16.6±2.9 days for tumors treated with RF alone, 15.5±2.1 days for tumors treated with liposomal quercetin alone, and 22.0±3.9 days with combined RF and quercetin. Additionally, combination quercetin/RF/doxorubicin therapy resulted in the longest survival (48.3±20.4 days), followed by RF/doxorubicin (29.9±3.8 days). CONCLUSIONS: IV liposomal quercetin in combination with RF ablation reduces tumor growth rates and improves animal endpoint survival. Further increases in endpoint survival can be seen by adding an additional anti-tumor adjuvant agent liposomal doxorubicin. This suggests that targeting several post-ablation processes with multi-drug nanotherapies can increase overall ablation efficacy.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Catheter Ablation , Heat-Shock Proteins/antagonists & inhibitors , Mammary Neoplasms, Experimental/therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Combined Modality Therapy , Doxorubicin/administration & dosage , Endpoint Determination , Female , Kaplan-Meier Estimate , Liposomes , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Quercetin/administration & dosage , Rats , Time Factors
20.
Pharm Res ; 29(2): 375-83, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21845505

ABSTRACT

PURPOSE: To evaluate and compare anticancer therapeutic effect of palmitoyl ascorbate liposomes (PAL) and free ascorbic acid (AA). METHODS: Liposomes incorporating palmitoyl ascorbate (PA) were prepared and evaluated for PA content by HPLC. To elucidate mechanism of action of cell death in vitro, effect of various H(2)O(2) scavengers and metal chelators on PA-mediated cytotoxicity was studied. Effect of various combinations of PAL and free AA on in vitro cytotoxicity was evaluated on 4T1 cells. In vivo, PAL formulation was modified with polyethylene glycol; effect of PEGylation on in vitro cytotoxicity was evaluated. Biodistribution of PEG-PAL formulation was investigated in female Balb/c mice bearing murine mammary carcinoma (4T1 cells). In vivo anticancer activity of PEG-PAL (PEG-PAL equivalent to 20 mg/kg of PA injected intravenously on alternate days) was compared with free AA therapy in same model. RESULTS: PEG-PAL treatment was significantly more effective than free AA treatment in slowing tumor growth. CONCLUSIONS: Nanoparticle formulations incorporating PA can kill cancer cells in vitro. The mechanism of PA cytotoxicity is based on production of extracellular reactive oxygen species and involves intracellular transition metals.


Subject(s)
Antioxidants/administration & dosage , Antioxidants/therapeutic use , Ascorbic Acid/analogs & derivatives , Ascorbic Acid/administration & dosage , Ascorbic Acid/therapeutic use , Mammary Neoplasms, Animal/drug therapy , Animals , Breast/drug effects , Breast/pathology , Cell Line, Tumor , Female , Liposomes , Lipoylation , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...