Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Biomacromolecules ; 21(8): 3038-3046, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32484665

ABSTRACT

Rapalogues are a unique class of drugs with both cytostatic and immunosuppressive properties. Two founding members, rapamycin (Rapa) and its chemical derivative everolimus (Eve), are extremely potent, but their clinical use presents multiple challenges. Being water-insoluble, administration is restricted to the oral route, which results in a low bioavailability of <10%. Human studies of rapalogues are reported to yield a high blood to plasma ratio and poor correlation between blood concentration and dose. Moreover, treatment results in dose-limiting toxicities such as stomatitis and pneumonitis, which often leads to discontinuation of therapy. We previously reported an elastin-like polypeptide decorated with two-headed FKBP rapalogue-binding domains. Called "FAF", this biomacromolecular drug-carrier solubilizes, retargets, and releases rapalogues within disease sites. FAF-rapalogue formulations are free of cosolvents or surfactants, which promotes their parenteral administration. In this study, subcutaneously given FAF-Rapa significantly suppressed tumor growth in a mouse model of hormone receptor positive (HR+) breast cancer, compared to an oral formulation of Eve (Affinitor). Additionally, mTOR, the pharmacological target of rapalogues, was inhibited to a greater extent in tumors of FAF-Rapa and FAF-Eve groups compared to mice that received oral Eve. No signaling suppression was detected in the liver and spleen, which were evaluated to represent off-target organs exposed to the circulating formulation.


Subject(s)
Breast Neoplasms , Everolimus , Animals , Breast Neoplasms/drug therapy , Female , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Peptides , Sirolimus/pharmacology
2.
Biomacromolecules ; 21(3): 1091-1102, 2020 03 09.
Article in English | MEDLINE | ID: mdl-31927993

ABSTRACT

The clinical utility of rapamycin (Rapa) is limited by solubility, bioavailability, and side effects. To overcome this, our team recently reported an elastin-like polypeptide (ELP) nanoparticle with high affinity, noncovalent drug binding, and integrin-mediated cellular uptake. Given the scarcity of pharmacology/toxicology studies of ELP-based drug carriers, this article explores safety and efficacy of ELP-Rapa. ELP-Rapa nanoparticles tested negative for hemolysis, did not interfere in plasma coagulation nor in platelet function, and did not activate the complement. Upon incubation with HepG2 cells, ELP-Rapa revealed significant cellular uptake and trafficking to acidic organelles, consistent with lysosomes. Internalized ELP-Rapa nanoparticles increased oxidative stress 4-fold compared to free drug or free ELP controls. However, mice bearing orthotopic hormone receptor positive BT-474 breast tumors, given a high dose (∼10-fold above therapeutic dose) of 1 month administration of ELP-Rapa, did not induce hepatotoxicity. On the other hand, tumor growth and mTOR signaling were suppressed without affecting body weight. Nanoparticles assembled using ELP technology appear to be a safe and efficient strategy for delivering Rapa.


Subject(s)
Breast Neoplasms , Elastin , Animals , Breast Neoplasms/drug therapy , Drug Carriers/therapeutic use , Elastin/therapeutic use , Female , Humans , Mice , Peptides/therapeutic use , Sirolimus/pharmacology
3.
Nanomedicine ; 24: 102111, 2020 02.
Article in English | MEDLINE | ID: mdl-31655204

ABSTRACT

Humanin (HN) is a hydrophobic 24-amino acid peptide derived from mitochondrial DNA that modulates cellular responses to oxidative stress and protects human retinal pigment epithelium (RPE) cells from apoptosis. To solubilize HN, this report describes two genetically-encoded fusions between HN and elastin-like polypeptides (ELP). ELPs provide steric stabilization and/or thermo-responsive phase separation. Fusions were designed to either remain soluble or phase separate at the physiological temperature of the retina. Interestingly, the soluble fusion assembles stable colloids with a hydrodynamic radius of 39.1 nm at 37°C. As intended, the thermo-responsive fusion forms large coacervates (>1,000 nm) at 37°C. Both fusions bind human RPE cells and protect against oxidative stress-induction of apoptosis (TUNEL, caspase-3 activation). Their activity is mediated through STAT3; furthermore, STAT3 inhibition eliminates their protection. These findings suggest that HN polypeptides may facilitate cellular delivery of biodegradable nanoparticles with potential protection against age-related diseases, including macular degeneration.


Subject(s)
Elastin , Epithelial Cells/metabolism , Intracellular Signaling Peptides and Proteins , Nanoparticles/chemistry , Oxidative Stress/drug effects , Peptides , Retinal Pigment Epithelium/metabolism , Apoptosis/drug effects , Cells, Cultured , Elastin/chemistry , Elastin/pharmacology , Epithelial Cells/pathology , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/pharmacology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Peptides/chemistry , Peptides/pharmacology , Retinal Pigment Epithelium/pathology
4.
ACS Biomater Sci Eng ; 7(2): 428-440, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33455201

ABSTRACT

Elastin-like polypeptides (ELPs) are modular, stimuli-responsive materials that self-assemble into protein-rich microdomains in response to heating. By cloning ELPs to effector proteins, expressed intracellular fusions can even modulate cellular pathways. A critical step in engineering these fusions is to determine and control their intracellular phase transition temperature (Tt). To do so, this Method paper describes a simple live-cell imaging technique to estimate the Tt of non-fluorescent ELP fusion proteins by co-transfection with a fluorescent ELP marker. Intracellular microdomain formation can then be visualized in live cells through the co-assembly of the non-fluorescent and fluorescent ELP fusion proteins. If the two ELP fusions have different Tt, the intracellular ELP mixture phase separates at the temperature corresponding to the fusion with the lower Tt. In addition, co-assembled ELP microdomains often exhibit pronounced differences in size or number, compared to single transfected treatments. These features enable live-cell imaging experiments and image analysis to determine the intracellular Tt of a library of related ELP fusions. As a case study, we employ the recently reported Caveolin1-ELP library (CAV1-ELPs). In addition to providing a detailed protocol, we also report the development of a useful FIJI plugin named SIAL (Simple Image Analysis Library), which contains programs for image randomization and blinding, phenotype scoring, and ROI selection. These tasks are important parts of the protocol detailed here and are also commonly employed in other image analysis workflows.


Subject(s)
Elastin , Peptides , Elastin/genetics , Peptides/genetics , Phase Transition , Temperature , Transition Temperature
5.
J Control Release ; 324: 317-329, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32428520

ABSTRACT

FLT3 receptor is an important therapeutic target in acute myeloid leukemia due to high incidence of mutations associated with poor clinical outcome. Targeted therapies against the FLT3 receptor, including small-molecule FLT3 tyrosine kinase inhibitors (TKIs) and anti-FLT3 antibodies, have demonstrated promising preclinical and even clinical efficacy. Yet, even with the current FDA approval for two FLT3 inhibitors, these modalities were unable to cure AML or significantly extend the lives of patients with a common mutation called FLT3-ITD. While FLT3 is a viable target, the approaches to inhibit its activity were inadequate. To develop a new modality for targeting FLT3, our team engineered an α-FLT3-A192 fusion protein composed of a single chain variable fragment antibody conjugated with an elastin-like polypeptide. These fusion proteins assemble into multi-valent nanoparticles with excellent stability and pharmacokinetic properties as well as in vitro and in vivo pharmacological activity in cellular and xenograft murine models of AML. In conclusion, α-FLT3-A192 fusions appear to be a viable new modality for targeting FLT3 in AML and warrant further preclinical development to bring it into the clinic.


Subject(s)
Leukemia, Myeloid, Acute , Nanoparticles , Animals , Elastin , Humans , Leukemia, Myeloid, Acute/drug therapy , Mice , Mutation , Protein Kinase Inhibitors , fms-Like Tyrosine Kinase 3/genetics
6.
ACS Biomater Sci Eng ; 6(1): 198-204, 2020 01 13.
Article in English | MEDLINE | ID: mdl-32542186

ABSTRACT

Caveolae are membrane organelles formed by submicron invaginations in the plasma membrane, and are involved in mechanosensing, cell signaling, and endocytosis. Although implicated broadly in physiology and pathophysiology, better tools are required to elucidate the precise role of caveolar processes through selective activation and inactivation of their trafficking. Our group recently reported that thermally-responsive elastin-like polypeptides (ELPs) can trigger formation of 'genetically engineered protein microdomains (GEPMs)' functionalized with either Clathrin-light chain or the epidermal growth factor receptor. This manuscript is the first report of this strategy to modulate caveolin-1 (CAV1). By attaching different ELP sequences to CAV1, mild heating can be used to self-assemble CAV1-ELP microdomains inside of cells. The temperature of self-assembly can be controlled by tuning the ELP sequence. The formation of CAV1-ELP microdomains internalizes Cholera Toxin Subunit B, a commonly used marker of caveolae mediated endocytosis. CAV1-ELPs also colocalize with Cavin 1, an essential component of functional caveolae biogenesis. With the emerging significance of caveolae in health and disease and the lack of specific probes to rapidly and reversibly affect caveolar function, CAV1-ELP microdomains are a new tool to rapidly probe caveolae associated processes in endocytosis, cell signaling, and mechanosensing.


Subject(s)
Caveolae , Caveolin 1 , Caveolae/metabolism , Caveolin 1/genetics , Elastin , Endocytosis , Temperature
7.
Front Pharmacol ; 9: 1184, 2018.
Article in English | MEDLINE | ID: mdl-30386244

ABSTRACT

Rapamycin (Rapa) is a highly potent drug; however, its clinical potential is limited by poor solubility, bioavailability, and cytotoxicity. To improve Rapa delivery, our team has fused the cognate protein receptor for Rapa, FKBP12, to high molecular weight elastin-like polypeptides (ELPs). One construct, FAF, includes an FKBP domain at each termini of an ELP. In a recent report, FAF/Rapa outperformed a family of related carriers with higher tumor accumulation and efficacy. Despite apparent efficacy, an explanation for how FAF carries Rapa into cells has not been elucidated. This manuscript explores the intracellular fate of FAF in MDA-MB-468, a triple negative (ER-/PR-/HER2-) breast cancer line. Based on a lack of displacement by excess unlabeled FAF, no evidence was found for the involvement of a receptor in cell-surface binding. Cellular association showed no dose-dependent saturation at concentrations up to 100 µM, which is consistent with uptake through fluid phase endocytosis. FAF does colocalize with dextran, a marker of fluid phase endocytosis. Upon internalization, both FAF and dextran target low pH intracellular compartments similarly. Despite likely exposure to lysosomal pH and proteolytic activity, intracellular FAF is eliminated from cells with a relatively long half-life of 17.7 and 19.0 h by confocal microscopy and SDS-PAGE respectively. A split luciferase reporter assay demonstrated that FAF delays the cytosolic access of Rapa in comparison to free drug by 30 min. A specific macropinocytosis inhibitor, amiloride, completely inhibits the cytosolic delivery of Rapa from FAF. Each of these results are consistent with macropinocytosis as the mechanism of cellular uptake necessary for the hand-off of Rapa from FKBP-based drug carriers like FAF to endogenous FKBP12 in the cytosol.

8.
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
SELECTION OF CITATIONS
SEARCH DETAIL