Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(25): e2201980119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35696571

ABSTRACT

Endosomal sorting maintains cellular homeostasis by recycling transmembrane proteins and associated proteins and lipids (termed "cargoes") from the endosomal network to multiple subcellular destinations, including retrograde traffic to the trans-Golgi network (TGN). Viral and bacterial pathogens subvert retrograde trafficking machinery to facilitate infectivity. Here, we develop a proteomic screen to identify retrograde cargo proteins of the endosomal SNX-BAR sorting complex promoting exit 1 (ESCPE-1). Using this methodology, we identify Neuropilin-1 (NRP1), a recently characterized host factor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, as a cargo directly bound and trafficked by ESCPE-1. ESCPE-1 mediates retrograde trafficking of engineered nanoparticles functionalized with the NRP1-interacting peptide of the SARS-CoV-2 spike (S) protein. CRISPR-Cas9 deletion of ESCPE-1 subunits reduces SARS-CoV-2 infection levels in cell culture. ESCPE-1 sorting of NRP1 may therefore play a role in the intracellular membrane trafficking of NRP1-interacting viruses such as SARS-CoV-2.


Subject(s)
COVID-19 , Endosomes , Host-Pathogen Interactions , Neuropilin-1 , SARS-CoV-2 , COVID-19/metabolism , COVID-19/virology , CRISPR-Cas Systems , Endosomes/virology , Gene Deletion , Humans , Nanoparticles , Neuropilin-1/genetics , Neuropilin-1/metabolism , Proteomics , SARS-CoV-2/metabolism , Sorting Nexins/metabolism , Spike Glycoprotein, Coronavirus/metabolism
2.
Proc Natl Acad Sci U S A ; 118(49)2021 11 16.
Article in English | MEDLINE | ID: mdl-34772761

ABSTRACT

Many phylogenetically distant animal viruses, including the new coronavirus severe acute respiratory syndrome coronavirus 2, have surface proteins with polybasic sites that are cleaved by host furin and furin-like proteases. Other than priming certain viral surface proteins for fusion, cleavage generates a carboxy-terminal RXXR sequence. This C-end Rule (CendR) motif is known to bind to neuropilin (NRP) receptors on the cell surface. NRPs are ubiquitously expressed, pleiotropic cell surface receptors with important roles in growth factor signaling, vascular biology, and neurobiology, as well as immune homeostasis and activation. The CendR-NRP receptor interaction promotes endocytic internalization and tissue spreading of different cargo, including viral particles. We propose that the interaction between viral surface proteins and NRPs plays an underappreciated and prevalent role in the transmission and pathogenesis of diverse viruses and represents a promising broad-spectrum antiviral target.


Subject(s)
COVID-19/virology , Neuropilins/metabolism , Virus Internalization , COVID-19/metabolism , Humans , Neuropilins/chemistry , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism
3.
Nucleic Acids Res ; 49(7): e38, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33444445

ABSTRACT

In vivo phage display is widely used for identification of organ- or disease-specific homing peptides. However, the current in vivo phage biopanning approaches fail to assess biodistribution of specific peptide phages across tissues during the screen, thus necessitating laborious and time-consuming post-screening validation studies on individual peptide phages. Here, we adopted bioinformatics tools used for RNA sequencing for analysis of high-throughput sequencing (HTS) data to estimate the representation of individual peptides during biopanning in vivo. The data from in vivo phage screen were analyzed using differential binding-relative representation of each peptide in the target organ versus in a panel of control organs. Application of this approach in a model study using low-diversity peptide T7 phage library with spiked-in brain homing phage demonstrated brain-specific differential binding of brain homing phage and resulted in identification of novel lung- and brain-specific homing peptides. Our study provides a broadly applicable approach to streamline in vivo peptide phage biopanning and to increase its reproducibility and success rate.


Subject(s)
Cell Surface Display Techniques/methods , High-Throughput Screening Assays/methods , Peptide Library , Peptides/metabolism , Animals , High-Throughput Nucleotide Sequencing/methods , Mice , Mice, Inbred BALB C , Tissue Distribution
4.
Mol Pharm ; 19(3): 895-903, 2022 03 07.
Article in English | MEDLINE | ID: mdl-35113575

ABSTRACT

Chronic lymphocytic leukemia (CLL) is the most common form of leukemia in adults. The disease is characterized by the accumulation of tumoral B cells resulting from a defect of apoptosis. We have in vitro and in vivo preclinically validated a tumor-penetrating peptide (named TT1) coupled to an interfering peptide (IP) that dissociates the interaction between the serine/threonine protein phosphatase 2A (PP2A) from its physiological inhibitor, the oncoprotein SET. This TT1-IP peptide has an antitumoral effect on CLL, as shown by the increased survival of mice bearing xenograft models of CLL, compared to control mice. The peptide did not show toxicity, as indicated by the mouse body weight and the biochemical parameters, such as renal and hepatic enzymes. In addition, the peptide-induced apoptosis in vitro of primary tumoral B cells isolated from CLL patients but not of those isolated from healthy patients. Finally, the peptide had approximately 5 h half-life in human serum and showed pharmacokinetic parameters compatible with clinical development as a therapeutic peptide against CLL.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Animals , Apoptosis , B-Lymphocytes/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mice , Peptides/pharmacology , Peptides/therapeutic use , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/pharmacology , Protein Phosphatase 2/therapeutic use
5.
Mol Ther ; 28(8): 1833-1845, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32497513

ABSTRACT

Systemic skin-selective therapeutics would be a major advancement in the treatment of diseases affecting the entire skin, such as recessive dystrophic epidermolysis bullosa (RDEB), which is caused by mutations in the COL7A1 gene and manifests in transforming growth factor-ß (TGF-ß)-driven fibrosis and malignant transformation. Homing peptides containing a C-terminal R/KXXR/K motif (C-end rule [CendR] sequence) activate an extravasation and tissue penetration pathway for tumor-specific drug delivery. We have previously described a homing peptide CRKDKC (CRK) that contains a cryptic CendR motif and homes to angiogenic blood vessels in wounds and tumors, but it cannot penetrate cells or tissues. In this study, we demonstrate that removal of the cysteine from CRK to expose the CendR sequence confers the peptide novel ability to home to normal skin. Fusion of the truncated CRK (tCRK) peptide to the C terminus of an extracellular matrix protein decorin (DCN), a natural TGF-ß inhibitor, resulted in a skin-homing therapeutic molecule (DCN-tCRK). Systemic DCN-tCRK administration in RDEB mice led to inhibition of TGF-ß signaling in the skin and significant improvement in the survival of RDEB mice. These results suggest that DCN-tCRK has the potential to be utilized as a novel therapeutic compound for the treatment of dermatological diseases such as RDEB.


Subject(s)
Epidermolysis Bullosa/etiology , Epidermolysis Bullosa/metabolism , Peptides/metabolism , Protein Interaction Domains and Motifs , Animals , Biomarkers , Disease Models, Animal , Epidermolysis Bullosa/pathology , Fibrosis , Immunohistochemistry , Mice , Mice, Knockout , Neuropilin-1/metabolism , Peptides/chemistry , Peptides/pharmacology , Protein Binding , Recombinant Fusion Proteins/pharmacology , Skin/drug effects , Skin/metabolism , Transforming Growth Factor beta/metabolism , Wound Healing/drug effects
6.
Adv Exp Med Biol ; 1295: 29-48, 2021.
Article in English | MEDLINE | ID: mdl-33543454

ABSTRACT

Tumor-homing peptides are widely used for improving tumor selectivity of anticancer drugs and imaging agents. The goal is to increase tumor uptake and reduce accumulation at nontarget sites. Here, we describe current approaches for tumor-homing peptide identification and validation, and provide comprehensive overview of classes of tumor-homing peptides undergoing preclinical and clinical development. We focus on unique mechanistic features and applications of a recently discovered class of tumor-homing peptides, tumor-penetrating C-end Rule (CendR) peptides, that can be used for tissue penetrative targeting of extravascular tumor tissue. Finally, we discuss unanswered questions and future directions in the field of development of peptide-guided smart drugs and imaging agents.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Delivery Systems , Humans , Neoplasms/drug therapy , Peptides/therapeutic use
7.
Angew Chem Int Ed Engl ; 60(31): 17018-17027, 2021 07 26.
Article in English | MEDLINE | ID: mdl-33908690

ABSTRACT

Novel anticancer compounds and their precision delivery systems are actively developed to create potent and well-tolerated anticancer therapeutics. Here, we report the synthesis of a novel anthracycline, Utorubicin (UTO), and its preclinical development as an anticancer payload for nanocarriers. Free UTO was significantly more toxic to cultured tumor cell lines than the clinically used anthracycline, doxorubicin. Nanoformulated UTO, encapsulated in polymeric nanovesicles (polymersomes, PS), reduced the viability of cultured malignant cells and this effect was potentiated by functionalization with a tumor-penetrating peptide (TPP). Systemic peptide-guided PS showed preferential accumulation in triple-negative breast tumor xenografts implanted in mice. At the same systemic UTO dose, the highest UTO accumulation in tumor tissue was seen for the TPP-targeted PS, followed by nontargeted PS, and free doxorubicin. Our study suggests potential applications for UTO in the treatment of malignant diseases and encourages further preclinical and clinical studies on UTO as a nanocarrier payload for precision cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/drug therapy , Mice , Molecular Conformation , Optical Imaging , Structure-Activity Relationship
8.
Mol Pharm ; 17(7): 2518-2531, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32421341

ABSTRACT

M2-like tumor-associated macrophages (M2 TAMs) play important roles in the resistance of tumors to immunotherapies. Selective depletion or reprogramming of M2 TAMs may sensitize the nonresponsive tumors for immune-mediated eradication. However, precision delivery of payloads to M2 TAMs, while sparing healthy tissues, has remained an unresolved challenge. Here, we studied the application of a short linear peptide (CSPGAK, "mUNO") for the delivery of molecular and nanoscale cargoes in M2 TAMs in vitro and the relevance of the peptide for in vivo targeting of early-stage primary breast tumors and metastatic lung foci. First, we performed in silico modeling and found that mUNO interacts with mouse CD206 via a binding site between lectin domains CTLD1 and CTLD2, the same site previously demonstrated to be involved in mUNO binding to human CD206. Second, we showed that cultured M2 macrophages take up fluorescein-labeled (FAM) polymersomes conjugated with mUNO using the sulfhydryl group of its N-terminal cysteine. Pulse/chase studies of FAM-mUNO in M2 macrophages suggested that the peptide avoided lysosomal entrapment and escaped from early endosomes. Third, our in vivo studies with FAM-mUNO demonstrated that intraperitoneal administration results in better pharmacokinetics and higher blood bioavailability than can be achieved with intravenous administration. Intraperitoneal FAM-mUNO, but not FAM-control, showed a robust accumulation in M2-skewed macrophages in mouse models of early primary breast tumor and lung metastasis. This targeting was specific, as no uptake was observed in nonmalignant control organs, including the liver, or other cell types in the tumor, including M1 macrophages. Collectively, our studies support the application of the CD206-binding mUNO peptide for delivery of molecular and nanoscale cargoes to M2 macrophages and manifest the relevance of this mode of targeting primary and metastatic breast tumors.


Subject(s)
Immunotherapy/methods , Lectins, C-Type/chemistry , Lung Neoplasms/diagnosis , Lymphatic Metastasis/diagnosis , Mannose-Binding Lectins/chemistry , Peptides/chemistry , Receptors, Cell Surface/chemistry , Triple Negative Breast Neoplasms/diagnosis , Tumor-Associated Macrophages/immunology , Animals , Binding Sites , Cell Differentiation , Cell Line, Tumor , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Carriers/pharmacokinetics , Female , Fluorescence , Humans , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymphatic Metastasis/diagnostic imaging , Lymphatic Metastasis/immunology , Lysosomes/metabolism , Maleimides/chemistry , Mannose Receptor , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Peptides/administration & dosage , Peptides/metabolism , Peptides/pharmacokinetics , Polyesters/chemistry , Polyethylene Glycols/chemistry , Polymers/administration & dosage , Polymers/chemistry , Polymers/pharmacology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Triple Negative Breast Neoplasms/diagnostic imaging , Triple Negative Breast Neoplasms/pathology , Tumor-Associated Macrophages/metabolism
9.
Mol Cell Probes ; 51: 101530, 2020 06.
Article in English | MEDLINE | ID: mdl-32035108

ABSTRACT

Using phage peptide library screening, we identified peptide-encoding phages that selectively home to the inflamed central nervous system (CNS) of mice with experimental autoimmune encephalomyelitis (EAE), a model of human multiple sclerosis (MS). A phage peptide display library encoding cyclic 9-amino-acid random peptides was first screened ex-vivo for binding to the CNS tissue of EAE mice, followed by in vivo screening in the diseased mice. Phage insert sequences that were present at a higher frequency in the CNS of EAE mice than in the normal (control) mice were identified by DNA sequencing. One of the phages selected in this manner, denoted as MS-1, was shown to selectively recognize CNS tissue in EAE mice. Individually cloned phages with this insert preferentially homed to EAE CNS after an intravenous injection. Similarly, systemically-administered fluorescence-labeled synthetic MS-1 peptide showed selective accumulation in the spinal cord of EAE mice. We suggest that peptide MS-1 might be useful for targeted drug delivery to CNS in EAE/MS.


Subject(s)
Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Peptides/metabolism , Animals , Brain/metabolism , Computational Biology , Encephalomyelitis, Autoimmune, Experimental/pathology , High-Throughput Nucleotide Sequencing , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Peptide Library , Peptides/genetics , Spinal Cord/metabolism
10.
Nano Lett ; 17(3): 1356-1364, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28178415

ABSTRACT

Antiangiogenic and vascular disrupting compounds have shown promise in cancer therapy, but tend to be only partially effective. We previously reported a potent theranostic nanosystem that was highly effective in glioblastoma and breast cancer mouse models, retarding tumor growth and producing some cures [ Agemy , L. et al. Proc. Natl. Acad. Sci. U.S.A. 2011 , 108 , 17450 - 17455 . Agemy , L. et al. Mol. Ther. 2013 , 21 , 2195 - 2204 .]. The nanosystem consists of iron oxide NPs ("nanoworms") coated with a composite peptide with tumor-homing and pro-apoptotic domains. The homing component targets tumor vessels by binding to p32/gC1qR at the surface or tumor endothelial cells. We sought to further improve the efficacy nanosystem by searching for an optimally effective homing peptide that would also incorporate a tumor-penetrating function. To this effect, we tested a panel of candidate p32 binding peptides with a sequence motif that conveys tumor-penetrating activity (CendR motif). We identified a peptide designated as Linear TT1 (Lin TT1) (sequence: AKRGARSTA) as most effective in causing tumor homing and penetration of the nanosystem. This peptide had the lowest affinity for p32 among the peptides tested. The low affinity may have moderated the avidity effect from the multivalent presentation on nanoparticles (NPs), such that the NPs avoid getting trapped by the so-called "binding-site barrier", which can hinder tissue penetration of compounds with a high affinity for their receptors. Treatment of breast cancer mice with the LinTT1 nanosystem showed greatly improved efficacy compared to the original system. These results identify a promising treatment modality and underscore the value of tumor penetration effect in improving the efficacy tumor treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Nanoparticles/therapeutic use , Peptides/therapeutic use , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Mice , Nanomedicine , Nanoparticles/chemistry , Nanoparticles/metabolism , Peptides/chemistry , Peptides/metabolism
11.
Molecules ; 23(5)2018 May 16.
Article in English | MEDLINE | ID: mdl-29772690

ABSTRACT

Over recent decades multiple therapeutic approaches have been explored for improved management of peritoneally disseminated malignancies-a grim condition known as peritoneal carcinomatosis (PC). Intraperitoneal (IP) administration can be used to achieve elevated local concentration and extended half-life of the drugs in the peritoneal cavity to improve their anticancer efficacy. However, IP-administered chemotherapeutics have a short residence time in the IP space, and are not tumor selective. An increasing body of work suggests that functionalization of drugs and nanoparticles with targeting peptides increases their peritoneal retention and provides a robust and specific tumor binding and penetration that translates into improved therapeutic response. Here we review the progress in affinity targeting of intraperitoneal anticancer compounds, imaging agents and nanoparticles with tumor-homing peptides. We review classes of tumor-homing peptides relevant for PC targeting, payloads for peptide-guided precision delivery, applications for targeted compounds, and the effects of nanoformulation of drugs and imaging agents on affinity-based tumor delivery.


Subject(s)
Peptides/administration & dosage , Peritoneal Neoplasms/diagnostic imaging , Peritoneal Neoplasms/drug therapy , Animals , Drug Delivery Systems , Humans , Injections, Intraperitoneal , Nanoparticles , Particle Size , Peptides/therapeutic use
12.
Tumour Biol ; 39(5): 1010428317701628, 2017 May.
Article in English | MEDLINE | ID: mdl-28468593

ABSTRACT

Peritoneal carcinomatosis results from dissemination of solid tumors in the peritoneal cavity, and is a common site of metastasis in patients with carcinomas of gastrointestinal or gynecological origin. Peritoneal carcinomatosis treatment is challenging as poorly vascularized, disseminated peritoneal micro-tumors are shielded from systemic anticancer drugs and drive tumor regrowth. Here, we describe the identification and validation of a tumor homing peptide CKRDLSRRC (IP3), which upon intraperitoneal administration delivers payloads to peritoneal metastases. IP3 peptide was identified by in vivo phage display on a mouse model of peritoneal carcinomatosis of gastric origin (MKN-45P), using high-throughput sequencing of the peptide-encoding region of phage genome as a readout. The IP3 peptide contains a hyaluronan-binding motif, and fluorescein-labeled IP3 peptide bound to immobilized hyaluronan in vitro. After intraperitoneal administration in mice bearing peritoneal metastases of gastric and colon origin, IP3 peptide homed robustly to macrophage-rich regions in peritoneal tumors, including poorly vascularized micro-tumors. Finally, we show that IP3 functionalization conferred silver nanoparticles the ability to home to peritoneal tumors of gastric and colonic origin, suggesting that it could facilitate targeted delivery of nanoscale payloads to peritoneal tumors. Collectively, our study suggests that the IP3 peptide has potential applications for targeting drugs, nanoparticles, and imaging agents to peritoneal tumors.


Subject(s)
Carcinoma/drug therapy , Hyaluronan Receptors/administration & dosage , Peptides/administration & dosage , Peritoneal Neoplasms/drug therapy , Animals , Bacteriophages/genetics , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Disease Models, Animal , Drug Delivery Systems , Humans , Hyaluronan Receptors/genetics , Mice , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neoplasm Metastasis , Peptides/genetics , Peritoneal Cavity/pathology , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/pathology
13.
Chembiochem ; 17(7): 570-5, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26895508

ABSTRACT

Cell surface p32, the target of LyP-1 homing peptide, is upregulated in tumors and atherosclerotic plaques and has been widely used as a receptor for systemic delivery of payloads. Here, we identified an improved LyP-1-mimicking peptide (TT1, CKRGARSTC). We used this peptide in a fluorescence polarization-based high-throughput screening of a 50,000-compound chemical library and identified a panel of compounds that bind p32 with low micromolar affinity. Among the hits identified in the screen, two compounds were shown to specifically bind to p32 in multiple assays. One of these compounds was chosen for an in vivo study. Nanoparticles surface-functionalized with this compound specifically adhered to surfaces coated with recombinant p32 and, when injected intravenously, homed to p32-expressing breast tumors in mice. This compound provides a lead for the development of p32-targeted affinity ligands that circumvent some of the limitations of peptide-based probes in guided drug delivery.


Subject(s)
Aminopyridines/chemistry , Breast Neoplasms/drug therapy , Drug Delivery Systems , Ethylenediamines/chemistry , Mitochondrial Proteins/administration & dosage , Peptides, Cyclic/administration & dosage , Aminopyridines/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Carrier Proteins , Cell Line, Tumor , Ethylenediamines/pharmacology , Female , Humans , Ligands , Mice , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Nanoparticles/chemistry
14.
Bioorg Med Chem Lett ; 26(6): 1618-1623, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26874401

ABSTRACT

Because nucleic acids (NAs) have immense potential value as therapeutics, the development of safe and effective synthetic NA vectors continues to attract much attention. In vivo applications of NA vectors require stabilized, nanometer-scale particles, but the commonly used approaches of steric stabilization with a polymer coat (e.g., PEGylation; PEG=poly(ethylene glycol)) interfere with attachment to cells, uptake, and endosomal escape. Conjugation of peptides to PEG-lipids can improve cell attachment and uptake for cationic liposome-DNA (CL-DNA) complexes. We present several synthetic approaches to peptide-PEG-lipids and discuss their merits and drawbacks. A lipid-PEG-amine building block served as the common key intermediate in all synthetic routes. Assembling the entire peptide-PEG-lipid by manual solid phase peptide synthesis (employing a lipid-PEG-carboxylic acid) allowed gram-scale synthesis but is mostly applicable to linear peptides connected via their N-terminus. Conjugation via thiol-maleimide or strain-promoted (copper-free) azide-alkyne cycloaddition chemistry is highly amenable to on-demand preparation of peptide-PEG-lipids, and the appropriate PEG-lipid precursors are available in a single chemical step from the lipid-PEG-amine building block. Azide-alkyne cycloaddition is especially suitable for disulfide-bridged peptides such as iRGD (cyclic CRGDKGPDC). Added at 10 mol% of a cationic/neutral lipid mixture, the peptide-PEG-lipids stabilize the size of CL-DNA complexes. They also affect cell attachment and uptake of nanoparticles in a peptide-dependent manner, thereby providing a platform for preparing stabilized, affinity-targeted CL-DNA nanoparticles.


Subject(s)
DNA/chemistry , Lipids/chemistry , Liposomes/chemistry , Peptides, Cyclic/chemical synthesis , Polyethylene Glycols/chemistry , Cations/chemistry , Humans , Liposomes/chemical synthesis , Molecular Structure , Nanoparticles/chemistry , Peptides, Cyclic/chemistry
15.
Proc Natl Acad Sci U S A ; 110(34): 13791-6, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23918357

ABSTRACT

Antibody cancer therapies rely on systemically accessible targets and suitable antibodies that exert a functional activity or deliver a payload to the tumor site. Here, we present proof-of-principle of in vivo selection of human antibodies in tumor-bearing mice that identified a tumor-specific antibody able to deliver a payload and unveils the target antigen. By using an ex vivo enrichment process against freshly disaggregated tumors to purge the repertoire, in combination with in vivo biopanning at optimized phage circulation time, we have identified a human domain antibody capable of mediating selective localization of phage to human prostate cancer xenografts. Affinity chromatography followed by mass spectrometry analysis showed that the antibody recognizes the proteasome activator complex PA28. The specificity of soluble antibody was confirmed by demonstrating its binding to the active human PA28αß complex. Whereas systemically administered control phage was confined in the lumen of blood vessels of both normal tissues and tumors, the selected phage spread from tumor vessels into the perivascular tumor parenchyma. In these areas, the selected phage partially colocalized with PA28 complex. Furthermore, we found that the expression of the α subunit of PA28 [proteasome activator complex subunit 1 (PSME1)] is elevated in primary and metastatic human prostate cancer and used anti-PSME1 antibodies to show that PSME1 is an accessible marker in mouse xenograft tumors. These results support the use of PA28 as a tumor marker and a potential target for therapeutic intervention in prostate cancer.


Subject(s)
Antibodies, Neoplasm/immunology , Biomarkers, Tumor/immunology , Immunotherapy/methods , Muscle Proteins/metabolism , Prostatic Neoplasms/immunology , Proteasome Endopeptidase Complex/metabolism , Animals , Antibodies, Neoplasm/metabolism , Antibody Specificity , Blotting, Western , Cell Surface Display Techniques , Chromatography, Affinity , Chromatography, Liquid , Drug Delivery Systems/methods , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Immunoprecipitation , Male , Mice , Mice, Nude , Prostatic Neoplasms/therapy , Statistics, Nonparametric , Tandem Mass Spectrometry
16.
Nat Mater ; 13(9): 904-11, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24907927

ABSTRACT

There is considerable interest in using nanoparticles as labels or to deliver drugs and other bioactive compounds to cells in vitro and in vivo. Fluorescent imaging, commonly used to study internalization and subcellular localization of nanoparticles, does not allow unequivocal distinction between cell surface-bound and internalized particles, as there is no methodology to turn particles 'off'. We have developed a simple technique to rapidly remove silver nanoparticles outside living cells, leaving only the internalized pool for imaging or quantification. The silver nanoparticle (AgNP) etching is based on the sensitivity of Ag to a hexacyanoferrate-thiosulphate redox-based destain solution. In demonstration of the technique we present a class of multicoloured plasmonic nanoprobes comprising dye-labelled AgNPs that are exceptionally bright and photostable, carry peptides as model targeting ligands, can be etched rapidly and with minimal toxicity in mice, and that show tumour uptake in vivo.


Subject(s)
Cells/metabolism , Metal Nanoparticles , Molecular Imaging/methods , Molecular Probes/chemistry , Molecular Probes/metabolism , Silver/chemistry , Silver/metabolism , Animals , Avidin/chemistry , Biological Transport , Cell Line, Tumor , Female , Humans , Mice , Molecular Probes/analysis , Molecular Probes/toxicity , Polyethylene Glycols/chemistry , Silver/toxicity
17.
Proc Natl Acad Sci U S A ; 108(17): 6909-14, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21486998

ABSTRACT

Affinity reagents that bind to specific molecular targets are an essential tool for both diagnostics and targeted therapeutics. There is a particular need for advanced technologies for the generation of reagents that specifically target cell-surface markers, because transmembrane proteins are notoriously difficult to express in recombinant form. We have previously shown that microfluidics offers many advantages for generating affinity reagents against purified protein targets, and we have now significantly extended this approach to achieve successful in vitro selection of T7 phage-displayed peptides that recognize markers expressed on live, adherent cells within a microfluidic channel. As a model, we have targeted neuropilin-1 (NRP-1), a membrane-bound receptor expressed at the surface of human prostate carcinoma cells that plays central roles in angiogenesis, cell migration, and invasion. We show that, compared to conventional biopanning methods, microfluidic selection enables more efficient discovery of peptides with higher affinity and specificity by providing controllable and reproducible means for applying stringent selection conditions against minimal amounts of target cells without loss. Using our microfluidic system, we isolate peptide sequences with superior binding affinity and specificity relative to the well known NRP-1-binding RPARPAR peptide. As such microfluidic systems can be used with a wide range of biocombinatorial libraries and tissue types, we believe that our method represents an effective approach toward efficient biomarker discovery from patient samples.


Subject(s)
Bacteriophage T7/genetics , Microfluidic Analytical Techniques/methods , Neuropilin-1/antagonists & inhibitors , Peptide Library , Cell Line, Tumor , Humans , Neuropilin-1/genetics , Neuropilin-1/metabolism
18.
Int J Nanomedicine ; 19: 4857-4875, 2024.
Article in English | MEDLINE | ID: mdl-38828195

ABSTRACT

Brain diseases are the most devastating problem among the world's increasingly aging population, and the number of patients with neurological diseases is expected to increase in the future. Although methods for delivering drugs to the brain have advanced significantly, none of these approaches provide satisfactory results for the treatment of brain diseases. This remains a challenge due to the unique anatomy and physiology of the brain, including tight regulation and limited access of substances across the blood-brain barrier. Nanoparticles are considered an ideal drug delivery system to hard-to-reach organs such as the brain. The development of new drugs and new nanomaterial-based brain treatments has opened various opportunities for scientists to develop brain-specific delivery systems that could improve treatment outcomes for patients with brain disorders such as Alzheimer's disease, Parkinson's disease, stroke and brain tumors. In this review, we discuss noteworthy literature that examines recent developments in brain-targeted nanomedicines used in the treatment of neurological diseases.


Subject(s)
Blood-Brain Barrier , Brain , Drug Delivery Systems , Nanomedicine , Humans , Nanomedicine/methods , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Drug Delivery Systems/methods , Animals , Nanoparticles/chemistry , Brain Diseases/drug therapy , Nanoparticle Drug Delivery System/chemistry , Nanoparticle Drug Delivery System/pharmacokinetics , Parkinson Disease/drug therapy , Alzheimer Disease/drug therapy
19.
Article in English | MEDLINE | ID: mdl-39012578

ABSTRACT

To achieve precision and selectivity, anticancer compounds and nanoparticles (NPs) can be targeted with affinity ligands that engage with malignancy-associated molecules in the blood vessels. While tumor-penetrating C-end Rule (CendR) peptides hold promise for precision tumor delivery, C-terminally exposed CendR peptides can accumulate undesirably in non-malignant tissues expressing neuropilin-1 (NRP-1), such as the lungs. One example of such promiscuous peptides is PL3 (sequence: AGRGRLVR), a peptide that engages with NRP-1 through its C-terminal CendR element, RLVR.Here, we report the development of PL3 derivatives that bind to NRP-1 only after proteolytic processing by urokinase-type plasminogen activator (uPA), while maintaining binding to the other receptor of the peptide, the C-domain of tenascin-C (TNC-C). Through a rational design approach and screening of a uPA-treated peptide-phage library (PL3 peptide followed by four random amino acids) on the recombinant NRP-1, derivatives of the PL3 peptide capable of binding to NRP-1 only post-uPA processing were successfully identified. In vitro cleavage, binding, and internalization assays, along with in vivo biodistribution studies in orthotopic glioblastoma-bearing mice, confirmed the efficacy of two novel peptides, PL3uCendR (AGRGRLVR↓SAGGSVA) and SKLG (AGRGRLVR↓SKLG), which exhibit uPA-dependent binding to NRP-1, reducing off-target binding to healthy NRP-1-expressing tissues. Our study not only unveils novel uPA-dependent TNC-C targeting CendR peptides but also introduces a broader paradigm and establishes a technology for screening proteolytically activated tumor-penetrating peptides.

20.
ACS Nano ; 18(22): 13983-13999, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38767983

ABSTRACT

In recent years, steady progress has been made in synthesizing and characterizing engineered nanoparticles, resulting in several approved drugs and multiple promising candidates in clinical trials. Regulatory agencies such as the Food and Drug Administration and the European Medicines Agency released important guidance documents facilitating nanoparticle-based drug product development, particularly in the context of liposomes and lipid-based carriers. Even with the progress achieved, it is clear that many barriers must still be overcome to accelerate translation into the clinic. At the recent conference workshop "Mechanisms and Barriers in Nanomedicine" in May 2023 in Colorado, U.S.A., leading experts discussed the formulation, physiological, immunological, regulatory, clinical, and educational barriers. This position paper invites open, unrestricted, nonproprietary discussion among senior faculty, young investigators, and students to trigger ideas and concepts to move the field forward.


Subject(s)
Nanomedicine , Humans , Drug Carriers/chemistry , Liposomes/chemistry , Nanoparticles/chemistry , United States
SELECTION OF CITATIONS
SEARCH DETAIL