Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters











Publication year range
1.
ACS Pharmacol Transl Sci ; 4(2): 589-612, 2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33860189

ABSTRACT

The safe and effective delivery of anticancer agents to diseased tissues is one of the significant challenges in cancer therapy. Conventional anticancer agents are generally cytotoxins with poor pharmacokinetics and bioavailability. Nanocarriers are nanosized particles designed for the selectivity of anticancer drugs and gene transport to tumors. They are small enough to extravasate into solid tumors, where they slowly release their therapeutic load by passive leakage or biodegradation. Using smart nanocarriers, the rate of release of the entrapped therapeutic(s) can be increased, and greater exposure of the tumor cells to the therapeutics can be achieved when the nanocarriers are exposed to certain internally (enzymes, pH, and temperature) or externally (light, magnetic field, and ultrasound) applied stimuli that trigger the release of their load in a safe and controlled manner, spatially and temporally. This review gives a comprehensive overview of recent research findings on the different types of stimuli-responsive nanocarriers and their application in cancer treatment with a particular focus on ultrasound.

3.
J Control Release ; 170(2): 233-41, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23714122

ABSTRACT

Molecular targeting of drug delivery nanocarriers is expected to improve their therapeutic index while decreasing their toxicity. Here we report the identification and characterization of novel peptide ligands specific for cells present in high-risk neuroblastoma (NB), a childhood tumor mostly refractory to current therapies. To isolate such targeting moieties, we performed combined in vitro/ex-vivo phage display screenings on NB cell lines and on tumors derived from orthotopic mouse models of human NB. By designing proper subtractive protocols, we identified phage clones specific either for the primary tumor, its metastases, or for their respective stromal components. Globally, we isolated 121 phage-displayed NB-binding peptides: 26 bound the primary tumor, 15 the metastatic mass, 57 and 23 their respective microenvironments. Of these, five phage clones were further validated for their specific binding ex-vivo to biopsies from stage IV NB patients and to NB tumors derived from mice. All five clones also targeted tumor cells and vasculature in vivo when injected into NB-bearing mice. Coupling of the corresponding targeting peptides with doxorubicin-loaded liposomes led to a significant inhibition in tumor volume and enhanced survival in preclinical NB models, thereby paving the way to their clinical development.


Subject(s)
Doxorubicin/administration & dosage , Nanoparticles/administration & dosage , Neuroblastoma/drug therapy , Peptides/administration & dosage , Animals , Cell Line, Tumor , Cell Surface Display Techniques , Cell Survival/drug effects , Cells, Cultured , Doxorubicin/chemistry , Female , Human Umbilical Vein Endothelial Cells , Humans , Liposomes , Mice , Mice, Nude , Nanoparticles/chemistry , Neuroblastoma/pathology , Peptides/chemistry , Peptides/pharmacokinetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
PLoS One ; 8(4): e62349, 2013.
Article in English | MEDLINE | ID: mdl-23626804

ABSTRACT

PURPOSE: To investigate the use of liposomal irinotecan (Irinophore C™) plus or minus 5-fluorouracil (5-FU) for the treatment of colorectal cancer. EXPERIMENTAL DESIGN: The effect of irinotecan (IRI) and/or 5-FU exposure times on cytotoxicity was assessed in vitro against HT-29 or LS174T human colon carcinoma cells. The pharmacokinetics and biodistribution of Irinophore C™ (IrC™) and 5-FU, administered alone or in combination, were compared in vivo. A subcutaneous model of HT-29 human colorectal cancer in Rag2-M mice was utilized to assess the efficacy of IrC™ alone, and in combination with 5-FU. RESULTS: The cytotoxicity of IRI and 5-FU were strongly dependent on exposure time. Synergistic interactions were observed following prolonged exposure to IRI/5-FU combinations. Pharmacokinetics/biodistribution studies demonstrated that the 5-FU elimination rate was decreased significantly when 5-FU was co-administered intravenously with IrC™, versus alone. Significant decreases in 5-FU elimination were also observed in plasma, with an associated increase of 5-FU in some tissues when 5-FU was given by intraperitoneal injection and IrC™ was given intravenously. The elimination of IrC™ was not significantly different when administered alone or in combination with 5-FU. Therapeutic studies demonstrated that single agent IrC™ was significantly more effective than the combination of IRI/5-FU; surprisingly, IrC™/5-FU combinations were no more effective than IrC™ alone. The administration of combinations of 5-FU (16 mg/kg) and IrC™ (60 mg IRI/kg) showed increased toxicity when compared to IrC™ alone. Treatment with IrC™ alone (60 mg IRI/kg) delayed the time required for a 5-fold increase in initial tumor volume to day 49, compared to day 23 for controls. When IrC™ (40 mg IRI/kg) was used in combination with 5-FU (16 mg/kg), the time to increase tumor volume 5-fold was 43 days, which was comparable to that achieved when using IrC™ alone (40 mg IRI/kg). CONCLUSIONS: Single agent IrC™ was well tolerated and has significant therapeutic potential. IrC™ may be a suitable replacement for IRI treatment, but its use with free 5-FU is complicated by IrC™-engendered changes in 5-FU pharmacokinetics/biodistribution which are associated with increased toxicity when using the combination.


Subject(s)
Camptothecin/analogs & derivatives , Colorectal Neoplasms/pathology , Fluorouracil/pharmacology , Animals , Camptothecin/administration & dosage , Camptothecin/pharmacology , Camptothecin/toxicity , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Disease Models, Animal , Drug Combinations , Female , Fluorouracil/administration & dosage , Fluorouracil/toxicity , HT29 Cells , Humans , Irinotecan , Liposomes , Male , Mice , Tumor Burden/drug effects
5.
J Drug Target ; 21(1): 87-96, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23039213

ABSTRACT

PURPOSE: We used two ligand-modified liposomal drugs to selectively deliver two different chemotherapeutics to tumor cells (TC) and tumor vasculature endothelial (TV) cells, and examined the therapeutic effect of altering the order of treatment administration, and the effect of the temporal spacing of the treatments on the accumulation of a second dose of liposomes and therapeutic activity. METHODS: Studies were completed in an orthotopic mouse model of human epidermal growth factor receptor 2 (HER2)-positive breast cancer, utilizing liposomal doxorubicin, targeted to TC via αHER2 Fab' fragments, and liposomal vincristine, targeted to CD13 on TV cells via NGR peptides. RESULTS AND DISCUSSION: Combination treatment with TV-targeted plus TC-targeted therapies was therapeutically superior to either single agent; switching the order of administration of the combination did not alter treatment efficacy. The tumor accumulation of a second dose of liposomes was increased if administered at 4 days after pre-treatment with TV-targeted therapy. Using a treatment schedule exploiting this increase, the dose of simultaneously administered combination therapy was halved without compromising therapeutic effect. CONCLUSION: Proof-of-concept studies revealed the therapeutic potential of a dual-targeted two drug approach against HER2-positive breast cancer, and may be applicable to the treatment of other solid tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Drug Delivery Systems , Mammary Neoplasms, Experimental/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/pathology , CD13 Antigens/metabolism , Doxorubicin/administration & dosage , Drug Administration Schedule , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Humans , Liposomes , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, SCID , Receptor, ErbB-2/metabolism , Vincristine/administration & dosage
6.
Adv Drug Deliv Rev ; 65(1): 36-48, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23036225

ABSTRACT

The first closed bilayer phospholipid systems, called liposomes, were described in 1965 and soon were proposed as drug delivery systems. The pioneering work of countless liposome researchers over almost 5 decades led to the development of important technical advances such as remote drug loading, extrusion for homogeneous size, long-circulating (PEGylated) liposomes, triggered release liposomes, liposomes containing nucleic acid polymers, ligand-targeted liposomes and liposomes containing combinations of drugs. These advances have led to numerous clinical trials in such diverse areas as the delivery of anti-cancer, anti-fungal and antibiotic drugs, the delivery of gene medicines, and the delivery of anesthetics and anti-inflammatory drugs. A number of liposomes (lipidic nanoparticles) are on the market, and many more are in the pipeline. Lipidic nanoparticles are the first nanomedicine delivery system to make the transition from concept to clinical application, and they are now an established technology platform with considerable clinical acceptance. We can look forward to many more clinical products in the future.


Subject(s)
Drug Delivery Systems/methods , Liposomes/administration & dosage , Liposomes/chemistry , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry , Animals , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Humans , Nanomedicine/methods , Nanoparticles/administration & dosage , Nanoparticles/chemistry
7.
Mol Pharm ; 9(11): 3266-76, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23030381

ABSTRACT

A family of 3-methoxypoly(ethylene glycol)-vinyl ether-1,2-dioleylglycerol (mPEG-VE-DOG) lipopolymer conjugates, designed on the basis of DFT calculations to possess a wide range of proton affinities, was synthesized and tested for their hydrolysis kinetics in neutral and acidic buffers. Extruded ∼100 nm liposomes containing these constructs in ≥90 mol % 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) produced dispersions that retained their calcein cargo for more than 2 days at pH 7.5, but released the encapsulated contents over a wide range of time scales as a function of the electronic properties of the vinyl ether linkage, the solution pH, and the mPEG-VE-DOG composition in the membrane. The in vivo performance of two different 90:10 DOPE:mPEG-VE-DOG compositions was also evaluated for blood circulation time and biodistribution in mice, using (125)I-tyraminylinulin as a label. The pharmacokinetic profiles gave a t(1/2) of 7 and 3 h for 90:10 DOPE:ST302 and 90:10 DOPE:ST502, respectively, with the liposomes being cleared predominantly by liver and spleen uptake. The behavior of these DOPE:mPEG-VE-DOG formulations is consistent with their relative rates of vinyl ether hydrolysis, i.e., the more acid-sensitive mPEG-VE-DOG derivatives produced faster leakage rates from DOPE:mPEG-VE-DOG liposomes, but decreased the blood circulation times in mice. These findings suggest that the vinyl ether-based PEG-lipid derivatives are promising agents for stabilizing acid-sensitive DOPE liposomes to produce formulations with a priori control over their pH responsiveness in vitro. Our data also suggest, however, that the same factors that contribute to enhanced acid sensitivity of the DOPE:mPEG-VE-DOG dispersions are also likely responsible for their reduced pharmacokinetic profiles.


Subject(s)
Diglycerides/pharmacokinetics , Lipids/chemical synthesis , Liposomes , Phosphatidylethanolamines/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Vinyl Compounds/pharmacokinetics , Acids/metabolism , Animals , Blood Circulation Time , Female , Hydrogen-Ion Concentration , Hydrolysis , Lipids/pharmacokinetics , Mice , Mice, Inbred BALB C , Tissue Distribution
8.
Mol Ther ; 19(12): 2201-12, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21829174

ABSTRACT

The anaplastic lymphoma kinase (ALK) is a tyrosine kinase receptor that is involved in the pathogenesis of different types of human cancers, including neuroblastoma (NB). In NB, ALK overexpression, or point mutations, are associated with poor prognosis and advanced stage disease. Inhibition of ALK kinase activity by small-molecule inhibitors in lung cancers carrying ALK translocations has shown therapeutic potential. However, secondary mutations may occur that, generate tumor resistance to ALK inhibitors. To overcome resistance to ALK inhibitors in NB, we adopted an alternative RNA interference (RNAi)-based therapeutic strategy that is able to knockdown ALK, regardless of its genetic status [mutated, amplified, wild-type (WT)]. NB cell lines, transduced by lentiviral short hairpin RNA (shRNA), showed reduced proliferation and increased apoptosis when ALK was knocked down. In mice, a nanodelivery system for ALK-specific small interfering RNA (siRNA), based on the conjugation of antibodies directed against the NB-selective marker GD(2) to liposomes, showed strong ALK knockdown in vivo in NB cells, which resulted in cell growth arrest, apoptosis, and prolonged survival. ALK knockdown was associated with marked reductions in vascular endothelial growth factor (VEGF) secretion, blood vessel density, and matrix metalloproteinases (MMPs) expression in vivo, suggesting a role for ALK in NB-induced neoangiogenesis and tumor invasion, confirming this gene as a fundamental oncogene in NB.


Subject(s)
Apoptosis , Mutation/genetics , Neovascularization, Pathologic/prevention & control , Neuroblastoma/blood supply , Neuroblastoma/therapy , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Anaplastic Lymphoma Kinase , Animals , Blotting, Western , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Gangliosides/immunology , Gangliosides/metabolism , HeLa Cells , Humans , Immunoenzyme Techniques , Liposomes , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Nude , Mice, SCID , Neuroblastoma/mortality , Phosphorylation , RNA Interference , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Survival Rate
9.
Mol Ther ; 19(6): 1131-40, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21487394

ABSTRACT

RNA interference molecules have some advantages as cancer therapeutics, including a proved efficacy on both wild-type (WT) and mutated transcripts and an extremely high sequence-specificity. The most significant hurdle to be overcome if exogenous small interfering RNAs (siRNA) is to be used therapeutically is the specific, effective, nontoxic delivery of siRNA to its intracellular site of action. At present, human applications are confined almost exclusively to targets within the liver, where the delivery systems naturally accumulate, and extra-hepatic targets remain a challenge. Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase that has recently been shown to contribute to the cell growth and progression of human neuroblastoma (NB). We investigated its potential as a therapeutic target in NB by generating anti-GD2-targeted nanoparticles that carry ALK-directed siRNA, which are specifically and efficiently delivered to GD2-expressing NB cells. Relative to free ALK-siRNA, anti-GD2-targeted liposomal formulations of ALK-siRNA had low plasma clearance, increased siRNA stability, and improved binding, uptake, silencing and induction of cell death, and specificity for NB cells. In NB xenografts, intravenous (i.v.) injection of the targeted ALK-siRNA liposomes showed gene-specific antitumor activity with no side effects. ALK-selective siRNA entrapped in anti-GD2-targeted nanoparticles is a promising new modality for NB treatment.


Subject(s)
Neuroblastoma/enzymology , Neuroblastoma/therapy , RNA, Small Interfering/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Anaplastic Lymphoma Kinase , Animals , Blotting, Western , Cell Line , Gene Silencing/physiology , Humans , Mice , Mice, Nude , Nanoparticles/chemistry , Neuroblastoma/genetics , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenograft Model Antitumor Assays
10.
J Control Release ; 145(1): 66-73, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20346382

ABSTRACT

The therapeutic index of anti-cancer drugs is increased when encapsulating them in tumor-targeted liposomes. Liposome-entrapped doxorubicin (DXR), targeting the tumor vasculature marker, aminopeptidase N (APN), displayed enhanced anti-tumor effects and prolonged survival in human neuroblastoma (NB)-bearing mice. Here we exploited a peptide ligand of aminopeptidase A (APA), discovered by phage display technology for delivery of liposomal DXR to perivascular tumor cells. Immunohistochemistry, performed in NB-bearing mice, showed APA expression in the vascular wall of NB primary and metastatic lesions. APA-targeted peptides displayed specific binding to APA-transfected cells in vitro, and also accumulation in the tumor of NB-bearing mice. Consequently, novel, APA-targeted, DXR-liposomes were developed and in vivo proof-of-principle was established, alone and in combination with APN-targeted DXR-loaded liposomes, in NB-bearing mice. Mice receiving APA-targeted liposomal DXR exhibited an increased life span in comparison to control mice, but to a lesser extent relative to that in mice treated with APN-targeted formulation, moreover the greatest increase in TUNEL-positive tumor cells was observed in animals treated with APN-targeted formulations. Mice treated with a combination of APA- and APN-targeted, liposomal DXR had a significant increase in life span compared to each treatment administered separately. There was a significant increase in the level of apoptosis in the tumors of mice on the combination therapy, and a pronounced destruction of the tumor vasculature with nearly total ablation of endothelial cells and pericytes. The availability of novel ligands binding to additional tumor vasculature-associated antigens will allow the design of sophisticated combinations of ligand-targeted liposomal anti-cancer drugs.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Doxorubicin/therapeutic use , Endothelial Cells/drug effects , Neuroblastoma/drug therapy , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , CD13 Antigens/biosynthesis , CD13 Antigens/chemistry , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Compounding , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Glutamyl Aminopeptidase/biosynthesis , Glutamyl Aminopeptidase/chemistry , Humans , Immunohistochemistry , Ligands , Liposomes , Mice , Mice, Nude , Neuroblastoma/blood supply , Neuroblastoma/metabolism , Neuroblastoma/pathology , Peptides/chemistry , Peptides/metabolism , Protein Binding , Xenograft Model Antitumor Assays
11.
Methods Enzymol ; 465: 225-49, 2009.
Article in English | MEDLINE | ID: mdl-19913170

ABSTRACT

Neuroblastoma (NB) is the most common extracranial solid tumor in childhood and the most frequently diagnosed neoplasm during infancy. Despite of aggressive treatment strategies, the 5-year survival rate for metastatic disease is still less than 60% and, consequently, novel therapeutic approaches are needed. For increasing the therapeutic index of anticancer drugs, while reducing side effects, one of the most promising strategies in modern chemotherapy is based on the development of innovative drug delivery systems, such as liposomes. "Anticancer drug"-loaded liposomes have demonstrated enhanced ability to target to the affected area, as well as increased antitumor efficacy compared to conventional drugs. Liposomes tend to extravasate preferentially and to accumulate into tumor interstitial fluids, due to the defective structure of the new angiogenic vessels within the tumor masses. This inherent tumor selectivity can be increased further by coupling tumor-specific antibodies or other targeting moieties to the surface of the lipid envelope. Here, we describe the methodology used in these studies, as well as the antitumor results obtained by the use of several "anticancer drugs," encapsulated into antibody- and peptide-targeted liposomal formulations, against NB.


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Fenretinide/therapeutic use , Liposomes , Neuroblastoma/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems , Fenretinide/administration & dosage , Gangliosides/chemistry , Gold , Mice , Microscopy, Electron, Transmission , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Neuroblastoma/blood supply
12.
Clin Cancer Res ; 14(22): 7320-9, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010847

ABSTRACT

PURPOSE: In vivo evaluation of good manufacturing practice-grade targeted liposomal doxorubicin (TVT-DOX), bound to a CD13 isoform expressed on the vasculature of solid tumors, in human tumor xenografts of neuroblastoma, ovarian cancer, and lung cancer. EXPERIMENTAL DESIGN: Mice were implanted with lung, ovarian, or neuroblastoma tumor cells via the pulmonary, peritoneal, or orthotopic (adrenal gland) routes, respectively, and treated, at different days post inoculation, with multiple doses of doxorubicin, administered either free or encapsulated in untargeted liposomes (Caelyx) or in TVT-DOX. The effect of TVT-DOX treatment on tumor cell proliferation, viability, apoptosis, and angiogenesis was studied by immunohistochemical analyses of neoplastic tissues and using the chick embryo chorioallantoic membrane assay. RESULTS: Compared with the three control groups (no doxorubicin, free doxorubicin, or Caelyx), statistically significant improvements in survival was seen in all three animal models following treatment with 5 mg/kg (maximum tolerated dose) of TVT-DOX, with long-term survivors occurring in the neuroblastoma group; increased survival was also seen at a dose of 1.7 mg/kg in mice bearing neuroblastoma or ovarian cancer. Minimal residual disease after surgical removal of neuroblastoma primary mass, and the enhanced response to TVT-DOX, was visualized and quantified by bioluminescence imaging and with magnetic resonance imaging. When treated with TVT-DOX, compared with Caelyx, all three tumor models, as assayed by immunohistochemistry and chorioallantoic membrane, showed statistically significant reductions in cell proliferation, blood vessel density, and microvessel area, showing increased cell apoptosis. CONCLUSION: TVT-DOX should be evaluated as a novel angiostatic strategy for adjuvant therapy of solid tumors.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , CD13 Antigens/metabolism , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Immunohistochemistry , Liposomes , Mice , Mice, Nude , Xenograft Model Antitumor Assays
13.
J Control Release ; 126(1): 85-94, 2008 Feb 18.
Article in English | MEDLINE | ID: mdl-18166243

ABSTRACT

Patients with advanced or metastatic melanoma have a very poor prognosis, due to the resistance of melanoma cells to conventional chemotherapy. We previously reported that coated cationic liposomes targeted with a monoclonal antibody against the disialoganglioside GD(2) and containing c-myc antisense oligodeoxynucleotides (alpha GD(2)-CCL[c-myc-as]) induced partial tumor growth arrest in melanoma xenografts. Here we addressed the role of c-myc-asODN treatment in the susceptibility to doxorubicin (DXR) in human melanoma cells. Cytotoxicity studies revealed that growth of melanoma cells was inhibited to a greater extent by alpha GD(2)-CCL[c-myc-as] than by the corresponding non-targeted formulations or by free c-myc-as. Targeted c-myc-as sensitized cells to DXR, reducing the IC(50) by approximately 10-fold. Scrambled ODNs had no effect on the IC(50) of DXR. Compared to either treatment alone, combination of targeted c-myc-as and DXR resulted in earlier apoptosis and in cell death after 2 days of treatment. In vivo experiments revealed that liposomal formulations of c-myc-as and DXR, both targeted via GD(2), led to the most pronounced delay in tumor growth when administered in a sequential manner. As a result, their combination translates into a statistically significant suppression of blood vessel density and an enhanced apoptosis, compared to all treatments given separately. Our data indicate the increasing cell sensitivity to DXR by c-myc-asODNs as a promising basis for developing novel anti-tumor strategy against advanced melanoma.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Antibodies, Monoclonal/therapeutic use , Doxorubicin/therapeutic use , Melanoma, Experimental/drug therapy , Oligodeoxyribonucleotides, Antisense/therapeutic use , Proto-Oncogene Proteins c-myc/biosynthesis , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Cell Adhesion , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Carriers/chemistry , Female , Gangliosides/immunology , Humans , Lipids/chemistry , Liposomes , Melanoma, Experimental/pathology , Mice , Mice, Nude , Neoplasm Transplantation , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/pharmacology
14.
J Control Release ; 126(1): 50-8, 2008 Feb 18.
Article in English | MEDLINE | ID: mdl-18068849

ABSTRACT

As part of an ongoing effort to develop a clinically acceptable doxorubicin formulation, targeted against B-cell malignancies, this study compared long-circulating (Stealth) immunoliposomes (SIL) that were targeted against the B-cell antigen CD19, via a whole HD37 monoclonal antibody (HD37 mAb), versus its Fab' fragment (HD37 Fab') or an HD37-c-myc-Cys-His5 single chain Fv fragment (scFv, HD37-CCH) directed against the same epitope. Compared to untargeted liposomes (SL), SIL showed increased binding in vitro to CD19-expressing Raji cells and, when loaded with doxorubicin (SIL-DXR), increased cytotoxicity against Raji (CD19(+)), but not Molt4 (CD19(-)) cells. Pharmacokinetics and biodistribution studies using dual-labeled liposomes (lipid and drug) in naive and Raji-bearing mice showed that SIL-DXR targeted via HD37 Fab' exhibited the same long circulation half-life as SL-DXR. SIL-DXR targeted via HD37-CCH was cleared faster than SL-DXR due to increased liver uptake, which was related to the poly-His and/or the c-myc tags in the scFv construct. SIL-DXR targeted via HD37 mAb was cleared rapidly from circulation due to Fc-mediated uptake in the liver and spleen. All three formulations of SIL-DXR extended the mean survival time of Raji-bearing mice compared to SL-DXR or free DXR. SIL-DXR targeted via HD37 Fab', which had the longest circulation half-life, appeared to be slightly more effective in prolonging survival times than SIL-DXR targeted via either HD37-CCH or HD37 mAb.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Antibodies, Monoclonal , Antigens, CD19/immunology , Burkitt Lymphoma/metabolism , Doxorubicin/administration & dosage , Immunoglobulin Fab Fragments , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/immunology , Cell Adhesion , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Female , Humans , Immunoglobulin Fab Fragments/administration & dosage , Immunoglobulin Fab Fragments/therapeutic use , Liposomes , Mice , Mice, Inbred BALB C , Mice, SCID , Nanoparticles , Neoplasm Transplantation , Tissue Distribution
15.
Mol Cancer Ther ; 6(11): 3019-27, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18025286

ABSTRACT

Site-specific delivery of anticancer agents to tumors represents a promising therapeutic strategy because it increases efficacy and reduces toxicity to normal tissues compared with untargeted drugs. Sterically stabilized immunoliposomes (SIL), guided by antibodies that specifically bind to well internalizing antigens on the tumor cell surface, are effective nanoscale delivery systems capable of accumulating large quantities of anticancer agents at the tumor site. The epithelial cell adhesion molecule (EpCAM) holds major promise as a target for antibody-based cancer therapy due to its abundant expression in many solid tumors and its limited distribution in normal tissues. We generated EpCAM-directed immunoliposomes by covalently coupling the humanized single-chain Fv antibody fragment 4D5MOCB to the surface of sterically stabilized liposomes loaded with the anticancer agent doxorubicin. In vitro, the doxorubicin-loaded immunoliposomes (SIL-Dox) showed efficient cell binding and internalization and were significantly more cytotoxic against EpCAM-positive tumor cells than nontargeted liposomes (SL-Dox). In athymic mice bearing established human tumor xenografts, pharmacokinetic and biodistribution analysis of SIL-Dox revealed long circulation times in the blood with a half-life of 11 h and effective time-dependent tumor localization, resulting in up to 15% injected dose per gram tissue. These favorable pharmacokinetic properties translated into potent antitumor activity, which resulted in significant growth inhibition (compared with control mice), and was more pronounced than that of doxorubicin alone and nontargeted SL-Dox at low, nontoxic doses. Our data show the promise of EpCAM-directed nanovesicular drug delivery for targeted therapy of solid tumors.


Subject(s)
Cell Adhesion Molecules/antagonists & inhibitors , Doxorubicin/pharmacology , Drug Delivery Systems , Epithelial Cells/metabolism , Nanotechnology/methods , Xenograft Model Antitumor Assays , Animals , Antigens, Neoplasm/metabolism , Biological Availability , Cell Adhesion Molecules/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cholesterol/analogs & derivatives , Cholesterol/metabolism , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Endocytosis/drug effects , Epithelial Cell Adhesion Molecule , Female , Humans , Liposomes , Mice
16.
Curr Med Chem ; 14(29): 3070-8, 2007.
Article in English | MEDLINE | ID: mdl-18220743

ABSTRACT

The central problem in cancer chemotherapy is the severe toxic side effects of anticancer drugs on healthy tissues. The use of liposomes as drug delivery vehicles for antitumour therapeutics has great potential to revolutionise the future of cancer therapy. As tumour architecture causes liposomes to preferentially accumulate at the tumour site, their use as drug carriers results in the localization of a greater amount of the loaded drug at the tumour site, thus improving cancer therapy and reducing the harmful non-specific side effects of chemotherapeutics. In addition, targeting of liposomal anticancer drugs to antigens expressed or over-expressed on tumour cells provides a very efficient system for increasing the therapeutic indices of the drugs. Animal models allow detailed examination of molecular and physiological basis of diseases and offer a frontline testing system for studying the involvement of specific genes and the efficacy of novel therapeutic approaches. Until recently, the most resorted experimental model of paediatric Neuroblastoma (NB) tumour is the subcutaneous xenograft in nude mice. However, the main disadvantage of this animal model is that it does not reflect the metastatic potential of NB cells, ultimately responsible for poor patient survival. A more realistic view of the clinical potential of targeted therapies could be obtained if a tumour model were available that better reflects the growth of advanced NB in children (i.e. large adrenal gland tumours and multiple small metastatic lesions). All current data support this concept and recommend that orthotopic implantation of tumour cells in recipient animals is mandatory for studies of tumour progression, angiogenesis, invasion, and metastasis. This review will focus on the description of the most clinically relevant animal models established to test the efficacy of targeted liposomal anti-tumour formulations for the treatment of Neuroblastoma.


Subject(s)
Antineoplastic Agents/administration & dosage , Neuroblastoma/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Drug Delivery Systems , Humans , Ligands , Liposomes , Neoplasm Transplantation , Neuroblastoma/immunology , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/therapeutic use , Transplantation, Heterologous
17.
Mol Cancer Ther ; 5(12): 3170-80, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17172421

ABSTRACT

Nanoscale drug delivery systems, such as sterically stabilized immunoliposomes binding to internalizing tumor-associated antigens, can increase therapeutic efficacy and reduce toxicity to normal tissues compared with nontargeted liposomes. The epithelial cell adhesion molecule (EpCAM) is of interest as a ligand for targeted drug delivery because it is abundantly expressed in solid tumors but shows limited distribution in normal tissues. To generate EpCAM-specific immunoliposomes for targeted cancer therapy, the humanized single-chain Fv antibody fragment 4D5MOCB was covalently linked to the exterior of coated cationic liposomes. As anticancer agent, we encapsulated the previously described antisense oligonucleotide 4625 specific for both bcl-2 and bcl-xL. The EpCAM-targeted immunoliposomes (SIL25) showed specific binding to EpCAM-overexpressing tumor cells, with a 10- to 20-fold increase in binding compared with nontargeted control liposomes. No enhanced binding was observed on EpCAM-negative control cells. On cell binding, SIL25 was efficiently internalized by receptor-mediated endocytosis, ultimately leading to down-regulation of both bcl-2 and bcl-xL expression on both the mRNA and protein level, which resulted in enhanced tumor cell apoptosis. In combination experiments, the use of SIL25 led to a 2- to 5-fold sensitization of EpCAM-positive tumor cells of diverse origin to death induction by doxorubicin. Our data show the promise of EpCAM-specific drug delivery systems, such as antisense-loaded immunoliposomes, for targeted cancer therapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Doxorubicin/pharmacology , Neoplasms/therapy , Oligonucleotides, Antisense/administration & dosage , Proto-Oncogene Proteins c-bcl-2/genetics , bcl-X Protein/genetics , Antigens, Neoplasm/genetics , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Down-Regulation , Drug Delivery Systems , Endocytosis/physiology , Epithelial Cell Adhesion Molecule , Female , Humans , Liposomes/administration & dosage , Liposomes/metabolism , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/therapy , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Oligonucleotides, Antisense/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/biosynthesis
18.
Anticancer Agents Med Chem ; 6(6): 513-23, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17100556

ABSTRACT

Nanoscale drug delivery systems (DDS) are used to circumvent some of the non-ideal properties of conventional anticancer chemotherapy drugs. Manipulation of the physical properties of DDS provides improved control over the pharmacokinetics (PK) and pharmacodynamics (PD) of the encapsulated drugs relative to free drugs. Liposomes are the archetypical nanoscale DDS and the first of these received clinical approval in 1990. DOXIL, liposomal doxorubicin, was the first commercially available liposomal anticancer drug (1995). It has an enhanced circulation half-life compared to the free drug because of its surface-grafted polyethylene glycol coating. DOXIL passively targets solid tumors, and once the liposomes localize in the tumor interstitial space, the cytotoxic drug is slowly released within the tumor. Liposomes can act as sustained release delivery system and manipulation of properties such as, liposome diameter, drug release rate, bioavailability and dosing schedule can significantly impact the therapeutic outcome of the liposomal drugs. This review will focus on how alteration of these properties can impact the therapeutic efficacy and side effect profiles of DDS.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/pharmacokinetics , Drug Delivery Systems , Liposomes , Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Carriers , Humans , Nanotechnology , Neoplasms/pathology , Polyethylene Glycols , Tissue Distribution
19.
Cancer Res ; 66(20): 10073-82, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17047071

ABSTRACT

Neuroblastoma, the most common solid tumor of infancy derived from the sympathetic nervous system, continues to present a formidable clinical challenge. Sterically stabilized immunoliposomes (SIL) have been shown to enhance the selective localization of entrapped drugs to solid tumors, with improvements in therapeutic indices. We showed that SIL loaded with doxorubicin (DXR) and targeted to the disialoganglioside receptor GD(2) [aGD(2)-SIL(DXR)] led to a selective inhibition of the metastatic growth of experimental models of human neuroblastoma. By coupling NGR peptides that target the angiogenic endothelial cell marker aminopeptidase N to the surface of DXR-loaded liposomes [NGR-SL(DXR)], we obtained tumor regression, pronounced destruction of the tumor vasculature, and prolonged survival of orthotopic neuroblastoma xenografts. Here, we showed good liposome stability, long circulation times, and enhanced time-dependent tumor accumulation of both the carrier and the drug. Antivascular effects against animal models of lung and ovarian cancer were shown for formulations of NGR-SL(DXR). In the chick embryo chorioallantoic assay, NGR-SL(DXR) substantially reduced the angiogenic potential of various neuroblastoma xenografts, with synergistic inhibition observed for the combination of NGR-SL(DXR) with aGD(2)-SIL(DXR). A significant improvement in antitumor effects was seen in neuroblastoma-bearing animal models when treated with the combined formulations compared with control mice or mice treated with either tumor- or vascular-targeted liposomal formulations, administered separately. The combined treatment resulted in a dramatic inhibition of tumor endothelial cell density. Long-term survivors were obtained only in animals treated with the combined tumor- and vascular-targeted formulations, confirming the pivotal role of combination therapies in treating aggressive metastatic neuroblastoma.


Subject(s)
Doxorubicin/administration & dosage , Myelin Proteins/metabolism , Neuroblastoma/blood supply , Neuroblastoma/drug therapy , Receptors, Cell Surface/metabolism , Animals , Apoptosis/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Female , GPI-Linked Proteins , Gangliosides/administration & dosage , Gangliosides/metabolism , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Nude , Mice, SCID , Myelin Proteins/administration & dosage , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neuroblastoma/metabolism , Nogo Receptor 1 , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacokinetics , Receptors, Cell Surface/administration & dosage , Tissue Distribution , Xenograft Model Antitumor Assays
20.
Cell Metab ; 3(5): 321-31, 2006 May.
Article in English | MEDLINE | ID: mdl-16679290

ABSTRACT

Phosphatidylcholine (PC) and phosphatidylethanolamine (PE) are major phospholipids in mammalian membranes. In liver, PC is synthesized via the choline pathway or by methylation of PE via phosphatidylethanolamine N-methyltransferase (PEMT). Pemt(-/-) mice fed a choline-deficient (CD) diet develop rapid steatohepatitis leading to liver failure. Steatosis is observed in CD mice that lack both PEMT and multiple drug-resistant protein 2 (MDR2), required for PC secretion into bile. We demonstrate that liver failure in CD-Pemt(-/-) mice is due to loss of membrane integrity caused by a decreased PC/PE ratio. The CD-Mdr2(-/-)/Pemt(-/-) mice escape liver failure by maintaining a normal PC/PE ratio. Manipulation of PC/PE levels suggests that this ratio is a key regulator of cell membrane integrity and plays a role in the progression of steatosis into steatohepatitis. The results have clinical implications as patients with nonalcoholic steatohepatitis have a decreased ratio of PC to PE compared to control livers.


Subject(s)
Cell Membrane/metabolism , Fatty Liver/metabolism , Liver Failure, Acute/metabolism , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Cell Membrane/pathology , Choline Deficiency/complications , Choline Deficiency/metabolism , Choline Deficiency/pathology , Disease Progression , Fatty Liver/etiology , Fatty Liver/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/ultrastructure , Liver Failure, Acute/etiology , Liver Failure, Acute/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylethanolamine N-Methyltransferase/genetics , Phosphatidylethanolamine N-Methyltransferase/metabolism , ATP-Binding Cassette Sub-Family B Member 4
SELECTION OF CITATIONS
SEARCH DETAIL