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 ; 33(3): 452-462, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35167278

ABSTRACT

Naturally occurring protein nanocages are promising drug carriers because of their uniform size and biocompatibility. Engineering efforts have enhanced the delivery properties of nanocages, but cell specificity and high drug loading remain major challenges. Herein, we fused the SpyTag peptide to the surface of engineered E2 nanocages to enable tunable nanocage decoration and effective E2 cell targeting using a variety of SpyCatcher (SC) fusion proteins. Additionally, the core of the E2 nanocage incorporated four phenylalanine mutations previously shown to allow hydrophobic loading of doxorubicin and pH-responsive release in acidic environments. We functionalized the surface of the nanocage with a highly cell-specific epidermal growth factor receptor (EGFR)-targeting protein conjugate, 4GE11-mCherry-SC, developed previously in our laboratories by employing unnatural amino acid (UAA) protein engineering chemistries. Herein, we demonstrated the benefits of this engineered protein nanocage construct for efficient drug loading, with a straightforward method for removal of the unloaded drug through elastin-like polypeptide-mediated inverse transition cycling. Additionally, we demonstrated approximately 3-fold higher doxorubicin internalization in inflammatory breast cancer cells compared to healthy breast epithelial cells, leading to targeted cell death at concentrations below the IC50 of free doxorubicin. Collectively, these results demonstrated the versatility of our UAA-based EGFR-targeting protein construct to deliver a variety of cargoes efficiently, including engineered E2 nanocages capable of site-specific functionalization and doxorubicin loading.


Subject(s)
Breast Neoplasms , Drug Carriers , Breast Neoplasms/drug therapy , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Delivery Systems/methods , ErbB Receptors , Female , Humans , Ligands
2.
Mol Pharm ; 19(2): 661-673, 2022 02 07.
Article in English | MEDLINE | ID: mdl-35040326

ABSTRACT

Intracellular delivery of protein therapeutics remains a significant challenge limiting the majority of clinically available protein drugs to extracellular targets. Strategies to deliver proteins to subcellular compartments have traditionally relied on cell-penetrating peptides, which can drive enhanced internalization but exhibit unreliable activity and are rarely able to target specific cells, leading to off-target effects. Moreover, few design rules exist regarding the relative efficacy of various endosomal escape strategies in proteins. Accordingly, we developed a simple fusion modification approach to incorporate endosomolytic peptides onto epidermal growth factor receptor (EGFR)-targeted protein conjugates and performed a systematic comparison of the endosomal escape efficacy, mechanism of action, and capacity to maintain EGFR-targeting specificity of conjugates modified with four different endosomolytic sequences of varying modes of action (Aurein 1.2, GALA, HA2, and L17E). Use of the recently developed Gal8-YFP assay indicated that the fusion of each endosomolytic peptide led to enhanced endosomal disruption. Additionally, the incorporation of each endosomolytic peptide increased the half-life of the internalized protein and lowered lysosomal colocalization, further supporting the membrane-disruptive capacity. Despite this, only EGFR-targeted conjugates modified with Aurein 1.2 or GALA maintained EGFR specificity. These results thus demonstrated that the choice of endosomal escape moiety can substantially affect targeting capability, cytotoxicity, and bioactivity and provided important new insights into endosomolytic peptide selection for the design of targeted protein delivery systems.


Subject(s)
Breast Neoplasms , Cell-Penetrating Peptides , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell-Penetrating Peptides/metabolism , Drug Delivery Systems/methods , Endosomes/metabolism , ErbB Receptors/metabolism , Female , Humans
3.
Curr Opin Biotechnol ; 71: 41-48, 2021 10.
Article in English | MEDLINE | ID: mdl-34157601

ABSTRACT

The importance of bioimaging and biosensing has been clear with the onset of the COVID-19 pandemic. In addition to viral detection, detection of tumors, glucose levels, and microbes is necessary for improved disease treatment and prevention. Bionanoparticles, such as extracellular vesicles and protein nanoparticles, are ideal platforms for biosensing and bioimaging applications because of their propensity for high density surface functionalization and large loading capacity. Scaffolding large numbers of sensing modules and detection modules onto bionanoparticles allows for enhanced analyte affinity and specificity as well as signal amplification for highly sensitive detection even at low analyte concentrations. Here we demonstrate the potential of bionanoparticles for bioimaging and biosensing by highlighting recent examples in literature that utilize protein nanoparticles and extracellular vesicles to generate highly sensitive detection devices with impressive signal amplification.


Subject(s)
Biosensing Techniques , COVID-19 , Nanoparticles , Humans , Pandemics , SARS-CoV-2
4.
Bioconjug Chem ; 31(10): 2272-2282, 2020 10 21.
Article in English | MEDLINE | ID: mdl-32931255

ABSTRACT

Proteins have the capacity to treat a multitude of diseases both as therapeutics and as drug carriers due to their complex functional properties, specificity toward binding partners, biocompatibility, and programmability. Despite this, native proteins often require assistance to target diseased tissue due to poor pharmacokinetic properties and membrane impermeability. Functionalizing therapeutic proteins and drug carriers through direct conjugation of delivery moieties can enhance delivery capabilities. Traditionally, this has been accomplished through bioconjugation methods that have little control over the location or orientation of the modification, leading to highly heterogeneous products with varying activity. A multitude of promising site-specific protein conjugation methods have been developed to allow more tailorable display of delivery moieties and thereby enhance protein activity, circulation properties, and targeting specificity. Here, we focus on three particularly promising site-specific bioconjugation techniques for protein delivery: unnatural amino acid incorporation, Sortase-mediated ligation, and SpyCatcher/SpyTag chemistry. In this review, we highlight the promise of site-specific bioconjugation for targeted drug delivery by summarizing impactful examples in literature, considering important design principles when constructing bioconjugates, and discussing our perspectives on future directions.


Subject(s)
Amino Acids/chemistry , Drug Carriers/chemistry , Proteins/administration & dosage , Amino Acids/chemical synthesis , Animals , Chemistry Techniques, Synthetic/methods , Drug Carriers/chemical synthesis , Drug Delivery Systems , Humans , Models, Molecular , Pharmaceutical Preparations/administration & dosage
5.
ACS Nano ; 14(10): 12642-12651, 2020 10 27.
Article in English | MEDLINE | ID: mdl-32924431

ABSTRACT

The hepatitis B virus-like particle (HBV VLP) is an attractive protein nanoparticle platform due to the availability of 240 modification sites for engineering purposes. Although direct protein insertion into the surface loop has been demonstrated, this decoration strategy is restricted by the size of the inserted protein moieties. Meanwhile, larger proteins can be decorated using chemical conjugations; yet these approaches perturb the integrity of more delicate proteins and can unfavorably orient the proteins, impairing active surface display. Herein, we aim to create a robust and highly modular method to produce smart HBV-based nanodevices by using the SpyCatcher/SpyTag system, which allows a wide range of peptides and proteins to be conjugated directly and simply onto the modified HBV capsids in a controlled and biocompatible manner. Our technology allows the modular surface modification of HBV VLPs with multiple components, which provides signal amplification, increased targeting avidity, and high therapeutic payload incorporation. We have achieved a yield of over 200 mg/L for these engineered HBV VLPs and demonstrated the flexibility of this platform in both biosensing and drug delivery applications. The ability to decorate over 200 nanoluciferases per VLP improved detection signal by over 1500-fold, such that low nanomolar levels of thrombin could be detected by the naked eye. Meanwhile, a dimeric prodrug-activating enzyme was loaded without cross-linking particles by coexpressing orthogonally labeled monomers. This along with a epidermal growth factor receptor-binding peptide enabled tunable uptake of HBV VLPs into inflammatory breast cancer cells, leading to efficient suicide enzyme delivery and cell killing.


Subject(s)
Hepatitis B , Pharmaceutical Preparations , Vaccines, Virus-Like Particle , Capsid , Hepatitis B virus , Humans , Peptides
6.
Bioconjug Chem ; 30(2): 432-442, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30615416

ABSTRACT

Proteins are ideal candidates for disease treatment because of their high specificity and potency. Despite this potential, delivery of proteins remains a significant challenge due to the intrinsic size, charge, and stability of proteins. Attempts to overcome these challenges have most commonly relied on direct conjugation of polymers and peptides to proteins via reactive groups on naturally occurring residues. While such approaches have shown some success, they allow limited control of the spacing and number of moieties coupled to proteins, which can hinder bioactivity and delivery capabilities of the therapeutic. Here, we describe a strategy to site-specifically conjugate delivery moieties to therapeutic proteins through unnatural amino acid (UAA) incorporation, in order to explore the effect of epidermal growth factor receptor (EGFR)-targeted ligand valency and spacing on internalization of proteins in EGFR-overexpressing inflammatory breast cancer (IBC) cells. Our results demonstrate the ability to enhance targeted protein delivery by tuning a small number of EGFR ligands per protein and clustering these ligands to promote multivalent ligand-receptor interactions. Furthermore, the tailorability of this simple approach was demonstrated through IBC-targeted cell death via the delivery of yeast cytosine deaminase (yCD), a prodrug converting enzyme.


Subject(s)
Amino Acids/metabolism , Cytosine Deaminase/administration & dosage , Luminescent Proteins/administration & dosage , Amino Acids/chemistry , Breast Neoplasms/metabolism , Cell Line, Tumor , Click Chemistry , Cytosine Deaminase/chemistry , Cytosine Deaminase/pharmacokinetics , Drug Delivery Systems , ErbB Receptors/metabolism , Female , Humans , Ligands , Luminescent Proteins/chemistry , Luminescent Proteins/pharmacokinetics , Models, Molecular , Protein Binding , Yeasts/enzymology , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...