Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Bioconjug Chem ; 28(11): 2715-2728, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28937754

ABSTRACT

Recombinant protein-polymer scaffolds such as elastin-like polypeptides (ELPs) offer drug-delivery opportunities including biocompatibility, monodispersity, and multifunctionality. We recently reported that the fusion of FK-506 binding protein 12 (FKBP) to an ELP nanoparticle (FSI) increases rapamycin (Rapa) solubility, suppresses tumor growth in breast cancer xenografts, and reduces side effects observed with free-drug controls. This new report significantly advances this carrier strategy by demonstrating the coassembly of two different ELP diblock copolymers containing drug-loading and tumor-targeting domains. A new ELP nanoparticle (ISR) was synthesized that includes the canonical integrin-targeting ligand (Arg-Gly-Asp, RGD). FSI and ISR mixed in a 1:1 molar ratio coassemble into bifunctional nanoparticles containing both the FKBP domain for Rapa loading and the RGD ligand for integrin binding. Coassembled nanoparticles were evaluated for bifunctionality by performing in vitro cell-binding and drug-retention assays and in vivo MDA-MB-468 breast tumor regression and tumor-accumulation studies. The bifunctional nanoparticle demonstrated superior cell target binding and similar drug retention to FSI; however, it enhanced the formulation potency, such that tumor growth was suppressed at a 3-fold lower dose compared to an untargeted FSI-Rapa control. This data suggests that ELP-mediated scaffolds are useful tools for generating multifunctional nanomedicines with potential activity in cancer.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Drug Carriers/chemistry , Elastin/chemistry , Integrins/metabolism , Sirolimus/administration & dosage , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Breast/drug effects , Breast/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Mice , Mice, Nude , Nanoparticles/chemistry , Peptides/chemistry , Sirolimus/pharmacokinetics , Sirolimus/pharmacology , Sirolimus/therapeutic use
2.
J Control Release ; 199: 156-67, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25481446

ABSTRACT

Millions of Americans suffer from dry eye disease, and there are few effective therapies capable of treating these patients. A decade ago, an abundant protein component of human tears was discovered and named lacritin (Lacrt). Lacrt has prosecretory activity in the lacrimal gland and mitogenic activity at the corneal epithelium. Similar to other proteins placed on the ocular surface, the durability of its effect is limited by rapid tear turnover. Motivated by the rationale that a thermo-responsive coacervate containing Lacrt would have better retention upon administration, we have constructed and tested the activity of a thermo-responsive Lacrt fused to an elastin-like polypeptide (ELP). Inspired from the human tropoelastin protein, ELP protein polymers reversibly phase separate into viscous coacervates above a tunable transition temperature. This fusion construct exhibited the prosecretory function of native Lacrt as illustrated by its ability to stimulate ß-hexosaminidase secretion from primary rabbit lacrimal gland acinar cells. It also increased tear secretion from non-obese diabetic (NOD) mice, a model of autoimmune dacryoadenitis, when administered via intra-lacrimal injection. Lacrt ELP fusion proteins undergo temperature-mediated assembly to form a depot inside the lacrimal gland. We propose that these Lacrt ELP fusion proteins represent a potential therapy for dry eye disease and the strategy of ELP-mediated phase separation may have applicability to other diseases of the ocular surface.


Subject(s)
Dry Eye Syndromes/drug therapy , Elastin/therapeutic use , Glycoproteins/therapeutic use , Proteins/therapeutic use , Actins/metabolism , Animals , Dacryocystitis/immunology , Delayed-Action Preparations , Elastin/chemistry , Female , Glycoproteins/chemistry , Hexosaminidase B/metabolism , Hot Temperature , Humans , In Vitro Techniques , Lacrimal Apparatus/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Primary Cell Culture , Proteins/chemistry , Rabbits , Recombinant Fusion Proteins , Tears/metabolism , Transcytosis
3.
ACS Nano ; 8(3): 2064-76, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24484356

ABSTRACT

B-cell lymphomas continue to occur with a high incidence. The chimeric antibody known as Rituximab (Rituxan) has become a vital therapy for these patients. Rituximab induces cell death via binding and clustering of the CD20 receptor by Fcγ expressing effector cells. Because of the limited mobility of effector cells, it may be advantageous to cluster CD20 directly using multivalent nanostructures. To explore this strategy, this manuscript introduces a nanoparticle that assembles from a fusion between a single chain antibody and a soluble protein polymer. These hybrid proteins express in Escherichia coli and do not require bioconjugation between the antibody and a substrate. Surprisingly a fusion between an anti-CD20 single chain antibody and a soluble protein polymer assemble worm-like nanostructures, which were characterized using light scattering and cryogenic transmission electron microscopy. These nanoworms competitively bind CD20 on two B-cell lymphoma cell lines, exhibit concentration-dependent induction of apoptosis, and induce apoptosis better than Rituximab alone. Similar activity was observed in vivo using a non-Hodgkin lymphoma xenograft model. In comparison to Rituximab, systemic nanoworms significantly slowed tumor growth. These findings suggest that hybrid nanoworms targeted at CD20 may be useful treatments for B-cell related malignancies. Because of the ubiquity of antibody therapeutics, related nanoworms may have uses against other molecular targets.


Subject(s)
Apoptosis/drug effects , Nanomedicine/methods , Nanoparticles/chemistry , Polymers/chemistry , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/pharmacology , Amino Acid Sequence , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Molecular Sequence Data , Protein Stability , Protein Structure, Secondary , Single-Chain Antibodies/pharmacokinetics , Temperature
4.
J Control Release ; 171(3): 330-8, 2013 Nov 10.
Article in English | MEDLINE | ID: mdl-23714121

ABSTRACT

Numerous nanocarriers of small molecules depend on either non-specific physical encapsulation or direct covalent linkage. In contrast, this manuscript explores an alternative encapsulation strategy based on high-specificity avidity between a small molecule drug and its cognate protein target fused to the corona of protein polymer nanoparticles. With the new strategy, the drug associates tightly to the carrier and releases slowly, which may decrease toxicity and promote tumor accumulation via the enhanced permeability and retention effect. To test this hypothesis, the drug Rapamycin (Rapa) was selected for its potent anti-proliferative properties, which give it immunosuppressant and anti-tumor activity. Despite its potency, Rapa has low solubility, low oral bioavailability, and rapid systemic clearance, which make it an excellent candidate for nanoparticulate drug delivery. To explore this approach, genetically engineered diblock copolymers were constructed from elastin-like polypeptides (ELPs) that assemble small (<100nm) nanoparticles. ELPs are protein polymers of the sequence (Val-Pro-Gly-Xaa-Gly)n, where the identity of Xaa and n determine their assembly properties. Initially, a screening assay for model drug encapsulation in ELP nanoparticles was developed, which showed that Rose Bengal and Rapa have high non-specific encapsulation in the core of ELP nanoparticles with a sequence where Xaa=Ile or Phe. While excellent at entrapping these drugs, their release was relatively fast (2.2h half-life) compared to their intended mean residence time in the human body. Having determined that Rapa can be non-specifically entrapped in the core of ELP nanoparticles, FK506 binding protein 12 (FKBP), which is the cognate protein target of Rapa, was genetically fused to the surface of these nanoparticles (FSI) to enhance their avidity towards Rapa. The fusion of FKBP to these nanoparticles slowed the terminal half-life of drug release to 57.8h. To determine if this class of drug carriers has potential applications in vivo, FSI/Rapa was administered to mice carrying a human breast cancer model (MDA-MB-468). Compared to free drug, FSI encapsulation significantly decreased gross toxicity and enhanced the anti-cancer activity. In conclusion, protein polymer nanoparticles decorated with the cognate receptor of a high potency, low solubility drug (Rapa) efficiently improved drug loading capacity and its release. This approach has applications to the delivery of Rapa and its analogs; furthermore, this strategy has broader applications in the encapsulation, targeting, and release of other potent small molecules.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Drug Carriers/chemistry , Elastin/chemistry , Nanoparticles/chemistry , Sirolimus/administration & dosage , Amino Acid Sequence , Animals , Antibiotics, Antineoplastic/therapeutic use , Breast/drug effects , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Molecular Sequence Data , Peptides/chemistry , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/chemistry
5.
Biomacromolecules ; 13(9): 2645-54, 2012 Sep 10.
Article in English | MEDLINE | ID: mdl-22849577

ABSTRACT

Peptide amphiphiles (PAs) self-assemble nanostructures with potential applications in drug delivery and tissue engineering. Some PAs share environmentally responsive behavior with their peptide components. Here we report a new type of PAs biologically inspired from human tropoelastin. Above a lower critical solution temperature (LCST), elastin-like polypeptides (ELPs) undergo a reversible inverse phase transition. Similar to other PAs, elastin-like PAs (ELPAs) assemble micelles with fiber-like nanostructures. Similar to ELPs, ELPAs have inverse phase transition behavior. Here we demonstrate control over the ELPAs fiber length and cellular uptake. In addition, we observed that both peptide assembly and nanofiber phase separation are accompanied by a distinctive secondary structure attributed primarily to a type-1 ß turn. We also demonstrate increased solubility of hydrophobic paclitaxel (PAX) in the presence of ELPAs. Due to their biodegradability, biocompatibility, and environmental responsiveness, elastin-inspired biopolymers are an emerging platform for drug and cell delivery; furthermore, the discovery of ELPAs may provide a new and useful approach to engineer these materials into stimuli-responsive gels and drug carriers.


Subject(s)
Biocompatible Materials/chemical synthesis , Drug Carriers/chemical synthesis , Nanofibers/chemistry , Peptides/chemical synthesis , Surface-Active Agents/chemical synthesis , Biocompatible Materials/pharmacology , Biomimetic Materials/chemistry , Cell Survival/drug effects , Drug Carriers/pharmacology , HEK293 Cells , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Micelles , Microscopy, Atomic Force , Nanofibers/ultrastructure , Paclitaxel/chemistry , Paclitaxel/pharmacology , Peptides/pharmacology , Phase Transition , Phosphatidylethanolamines/chemistry , Protein Structure, Secondary , Solubility , Surface-Active Agents/pharmacology , Temperature , Tropoelastin/chemistry , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
6.
Adv Drug Deliv Rev ; 61(11): 940-52, 2009 Sep 30.
Article in English | MEDLINE | ID: mdl-19628014

ABSTRACT

The tumor microenvironment provides multiple cues that may be exploited to improve the efficacy of established chemotherapeutics; furthermore, polypeptides are uniquely situated to capitalize on these signals. Peptides provide: 1) a rich repertoire of biologically specific interactions to draw upon; 2) environmentally responsive phase behaviors, which may be tuned to respond to signatures of disease; 3) opportunities to direct self-assembly; 4) control over routes of biodegradation; 5) the option to seamlessly combine functionalities into a single polymer via a one-step biosynthesis. As development of cancer-targeted nanocarriers expands, peptides provide a unique source of functional units that may target disease. This review explores potential microenvironmental physiology indicative of tumors and peptides that have demonstrated an ability to target and deliver to these signals.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers , Peptides/administration & dosage , Hydrogen-Ion Concentration , Ligands , Oxidation-Reduction , Permeability , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...