Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Pathol ; 245(2): 209-221, 2018 06.
Article in English | MEDLINE | ID: mdl-29603739

ABSTRACT

High-grade brain cancer such as glioblastoma (GBM) remains an incurable disease. A common feature of GBM is the angiogenic vasculature, which can be targeted with selected peptides for payload delivery. We assessed the ability of micelle-tagged, vascular homing peptides RGR, CGKRK and NGR to specifically bind to blood vessels in syngeneic orthotopic GBM models. By using the peptide CGKRK to deliver the tumour necrosis factor (TNF) superfamily member LIGHT (also known as TNF superfamily member 14; TNFSF14) to angiogenic tumour vessels, we have generated a reagent that normalizes the brain cancer vasculature by inducing pericyte contractility and re-establishing endothelial barrier integrity. LIGHT-mediated vascular remodelling also activates endothelia and induces intratumoural high endothelial venules (HEVs), which are specialized blood vessels for lymphocyte infiltration. Combining CGKRK-LIGHT with anti-vascular endothelial growth factor and checkpoint blockade amplified HEV frequency and T-cell accumulation in GBM, which is often sparsely infiltrated by immune effector cells, and reduced tumour burden. Furthermore, CGKRK and RGR peptides strongly bound to blood vessels in freshly resected human GBM, demonstrating shared peptide-binding activities in mouse and human primary brain tumour vessels. Thus, peptide-mediated LIGHT targeting is a highly translatable approach in primary brain cancer to reduce vascular leakiness and enhance immunotherapy. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Brain Neoplasms/drug therapy , Cell-Penetrating Peptides/metabolism , Drug Carriers , Glioblastoma/drug therapy , Neovascularization, Pathologic , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Vascular Remodeling/drug effects , Venules/drug effects , Adult , Aged , Aged, 80 and over , Angiogenesis Inhibitors/metabolism , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line , Drug Compounding , Female , Glioblastoma/blood , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Micelles , Middle Aged , Molecular Targeted Therapy , Pericytes/drug effects , Pericytes/metabolism , Pericytes/pathology , Phenotype , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Venules/metabolism , Venules/pathology
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Mol Pharm ; 11(12): 4374-84, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25333706

ABSTRACT

This study sought to develop a liposomal delivery system of fasudil--an investigational drug for the treatment of pulmonary arterial hypertension (PAH)--that will preferentially accumulate in the PAH lungs. Liposomal fasudil was prepared by film-hydration method, and the drug was encapsulated by active loading. The liposome surface was coated with a targeting moiety, CARSKNKDC, a cyclic peptide; the liposomes were characterized for size, polydispersity index, zeta potential, and storage and nebulization stability. The in vitro drug release profiles and uptake by TGF-ß activated pulmonary arterial smooth muscle cells (PASMC) and alveolar macrophages were evaluated. The pharmacokinetics were monitored in male Sprague-Dawley rats, and the pulmonary hemodynamics were studied in acute and chronic PAH rats. The size, polydispersity index (PDI), and zeta potential of the liposomes were 206-216 nm, 0.058-0.084, and -20-42.7 mV, respectively. The formulations showed minimal changes in structural integrity when nebulized with a commercial microsprayer. The optimized formulation was stable for >4 weeks when stored at 4 °C. Fasudil was released in a continuous fashion over 120 h with a cumulative release of 76%. Peptide-linked liposomes were taken up at a higher degree by TGF-ß activated PASMCs; but alveolar macrophages could not engulf peptide-coated liposomes. The formulations did not injure the lungs; the half-life of liposomal fasudil was 34-fold higher than that of plain fasudil after intravenous administration. Peptide-linked liposomal fasudil, as opposed to plain liposomes, reduced the mean pulmonary arterial pressure by 35-40%, without influencing the mean systemic arterial pressure. This study establishes that CAR-conjugated inhalable liposomal fasudil offers favorable pharmacokinetics and produces pulmonary vasculature specific dilatation.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Hypertension, Pulmonary/drug therapy , Liposomes/chemistry , Peptides/chemistry , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/chemistry , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Animals , Male , Rats , Rats, Sprague-Dawley , Vasodilator Agents/chemistry , Vasodilator Agents/therapeutic use
8.
Proc Natl Acad Sci U S A ; 108(42): 17450-5, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-21969599

ABSTRACT

Antiangiogenic therapy can produce transient tumor regression in glioblastoma (GBM), but no prolongation in patient survival has been achieved. We have constructed a nanosystem targeted to tumor vasculature that incorporates three elements: (i) a tumor-homing peptide that specifically delivers its payload to the mitochondria of tumor endothelial cells and tumor cells, (ii) conjugation of this homing peptide with a proapoptotic peptide that acts on mitochondria, and (iii) multivalent presentation on iron oxide nanoparticles, which enhances the proapoptotic activity. The iron oxide component of the nanoparticles enabled imaging of GBM tumors in mice. Systemic treatment of GBM-bearing mice with the nanoparticles eradicated most tumors in one GBM mouse model and significantly delayed tumor development in another. Coinjecting the nanoparticles with a tumor-penetrating peptide further enhanced the therapeutic effect. Both models used have proven completely resistant to other therapies, suggesting clinical potential of our nanosystem.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Oligopeptides/administration & dosage , Amino Acid Sequence , Angiogenesis Inhibitors/chemistry , Animals , Apoptosis/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/metabolism , Glioblastoma/pathology , Human Umbilical Vein Endothelial Cells , Humans , Magnetite Nanoparticles/administration & dosage , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Mitochondria/drug effects , Mitochondria/metabolism , Oligopeptides/chemistry
9.
J Comput Aided Mol Des ; 27(1): 31-43, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23239171

ABSTRACT

We present a chemical strategy to engineer analogs of the tumor-homing peptide CREKA (Cys-Arg-Glu-Lys-Ala), which binds to fibrin and fibrin-associated clotted plasma proteins in tumor vessels (Simberg et al. in Proc Natl Acad Sci USA 104:932-936, 2007) with improved ability to inhibit tumor growth. Computer modeling using a combination of simulated annealing and molecular dynamics were carried out to design targeted replacements aimed at enhancing the stability of the bioactive conformation of CREKA. Because this conformation presents a pocket-like shape with the charged groups of Arg, Glu and Lys pointing outward, non-proteinogenic amino acids α-methyl and N-methyl derivatives of Arg, Glu and Lys were selected, rationally designed and incorporated into CREKA analogs. The stabilization of the bioactive conformation predicted by the modeling for the different CREKA analogs matched the tumor fluorescence results, with tumor accumulation increasing with stabilization. Here we report the modeling, synthetic procedures, and new biological assays used to test the efficacy and utility of the analogs. Combined, our results show how studies based on multi-disciplinary collaboration can converge and lead to useful biomedical advances.


Subject(s)
Antineoplastic Agents/chemistry , Drug Design , Oligopeptides/chemistry , Amino Acids/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Mice , Molecular Dynamics Simulation , Nanostructures/chemistry , Oligopeptides/metabolism , Oligopeptides/pharmacology , Peptides/chemistry , Protein Conformation
10.
Proc Natl Acad Sci U S A ; 107(3): 981-6, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20080556

ABSTRACT

A significant barrier to the clinical translation of systemically administered therapeutic nanoparticles is their tendency to be removed from circulation by the mononuclear phagocyte system. The addition of a targeting ligand that selectively interacts with cancer cells can improve the therapeutic efficacy of nanomaterials, although these systems have met with only limited success. Here, we present a cooperative nanosystem consisting of two discrete nanomaterials. The first component is gold nanorod (NR) "activators" that populate the porous tumor vessels and act as photothermal antennas to specify tumor heating via remote near-infrared laser irradiation. We find that local tumor heating accelerates the recruitment of the second component: a targeted nanoparticle consisting of either magnetic nanoworms (NW) or doxorubicin-loaded liposomes (LP). The targeting species employed in this work is a cyclic nine-amino acid peptide LyP-1 (Cys-Gly-Asn-Lys-Arg-Thr-Arg-Gly-Cys) that binds to the stress-related protein, p32, which we find to be upregulated on the surface of tumor-associated cells upon thermal treatment. Mice containing xenografted MDA-MB-435 tumors that are treated with the combined NR/LyP-1LP therapeutic system display significant reductions in tumor volume compared with individual nanoparticles or untargeted cooperative system.


Subject(s)
Nanostructures , Neoplasms/therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Humans , Neoplasms/drug therapy
11.
Blood ; 116(15): 2847-56, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-20587786

ABSTRACT

The tumor-homing pentapeptide CREKA (Cys-Arg-Glu-Lys-Ala) specifically homes to tumors by binding to fibrin and fibrin-associated clotted plasma proteins in tumor vessels. Previous results show that CREKA-coated superparamagnetic iron oxide particles can cause additional clotting in tumor vessels, which creates more binding sites for the peptide. We have used this self-amplifying homing system to develop theranostic nanoparticles that simultaneously serve as an imaging agent and inhibit tumor growth by obstructing tumor circulation through blood clotting. The CREKA nanoparticles were combined with nanoparticles coated with another tumor-homing peptide, CRKDKC, and nanoparticles with an elongated shape (nanoworms) were used for improved binding efficacy. The efficacy of the CREKA peptide was then increased by replacing some residues with nonproteinogenic counterparts, which increased the stability of the peptide in the circulation. Treatment of mice bearing orthotopic human prostate cancer tumors with the targeted nanoworms caused extensive clotting in tumor vessels, whereas no clotting was observed in the vessels of normal tissues. Optical and magnetic resonance imaging confirmed tumor-specific targeting of the nanoworms, and ultrasound imaging showed reduced blood flow in tumor vessels. Treatment of mice with prostate cancer with multiple doses of the nanoworms induced tumor necrosis and a highly significant reduction in tumor growth.


Subject(s)
Metal Nanoparticles/therapeutic use , Oligopeptides/administration & dosage , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/therapy , Animals , Cell Line, Tumor , Drug Delivery Systems , Ferric Compounds/chemistry , Humans , Magnetic Resonance Imaging , Male , Metal Nanoparticles/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
12.
Proc Natl Acad Sci U S A ; 106(38): 16157-62, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19805273

ABSTRACT

Screening of phage libraries expressing random peptides for binding to prostate cancer cells primarily yielded peptides that had a C-terminal arginine (or rarely lysine) residue, usually in a consensus context R/KXXR/K. Phage expressing these sequences and synthetic nanoparticles coated with them bound to and were internalized into cells. The C-terminal arginine (or lysine) was essential to the activity; adding another amino acid, or even blocking the free carboxyl group of this arginine residue by amidation, eliminated the binding and internalizing activity. An internal R/KXXR/K can be exposed and switched on by a cleavage by a protease. The strict requirement for C-terminal exposure of the motif prompted us to term the phenomenon the C-end rule (CendR). Affinity chromatography showed that the CendR peptides bind to neuropilin-1 (NRP-1) on the target cells. NRP-1 is a cell-surface receptor that plays an essential role in angiogenesis, regulation of vascular permeability, and the development of the nervous system. VEGF-A165 and other ligands of NRP-1 possess a C-terminal CendR sequence that interacts with the b1 domain of NRP-1 and causes cellular internalization and vascular leakage. Our CendR peptides have similar effects, particularly when made multivalent through coupling to a particle. We also noted a unique and important activity of these peptides: penetration and transportation through tissues. The peptides were able to take payloads up to the nanoparticle size scale deep into extravascular tissue. Our observations have implications in drug delivery and penetration of tissue barriers and tumors.


Subject(s)
Arginine/metabolism , Neuropilin-1/metabolism , Peptides/metabolism , Prostatic Neoplasms/metabolism , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Cell Line, Tumor , Chromatography, Affinity , Endocytosis , Humans , Lung/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Confocal , Neoplasm Transplantation , Neuropilin-1/genetics , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Library , Peptides/chemistry , Peptides/pharmacokinetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Binding , Tissue Distribution , Transfection , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/metabolism
13.
Proc Natl Acad Sci U S A ; 106(24): 9815-9, 2009 Jun 16.
Article in English | MEDLINE | ID: mdl-19487682

ABSTRACT

Subtle clotting that occurs on the luminal surface of atherosclerotic plaques presents a novel target for nanoparticle-based diagnostics and therapeutics. We have developed modular multifunctional micelles that contain a targeting element, a fluorophore, and, when desired, a drug component in the same particle. Targeting atherosclerotic plaques in ApoE-null mice fed a high-fat diet was accomplished with the pentapeptide cysteine-arginine-glutamic acid-lysine-alanine, which binds to clotted plasma proteins. The fluorescent micelles bind to the entire surface of the plaque, and notably, concentrate at the shoulders of the plaque, a location that is prone to rupture. We also show that the targeted micelles deliver an increased concentration of the anticoagulant drug hirulog to the plaque compared with untargeted micelles.


Subject(s)
Atherosclerosis/pathology , Micelles , Animals , Aorta/pathology , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Fluorescent Antibody Technique , Mice , Mice, Knockout
14.
Pharmaceutics ; 13(10)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34683956

ABSTRACT

Diagnostic imaging of aggressive cancer with a high stroma content may benefit from the use of imaging contrast agents targeted with peptides that have high binding affinity to the extracellular matrix (ECM). In this study, we report the use of superparamagnetic iron-oxide nanoparticles (IO-NP) conjugated to a nonapeptide, CSGRRSSKC (CSG), which specifically binds to the laminin-nidogen-1 complex in tumours. We show that CSG-IO-NP accumulate in tumours, predominantly in the tumour ECM, following intravenous injection into a murine model of pancreatic neuroendocrine tumour (PNET). In contrast, a control untargeted IO-NP consistently show poor tumour uptake, and IO-NP conjugated to a pentapeptide. CREKA that bind fibrin clots in blood vessels show restricted uptake in the angiogenic vessels of the tumours. CSG-IO-NP show three-fold higher intratumoral accumulation compared to CREKA-IO-NP. Magnetic resonance imaging (MRI) T2-weighted scans and T2 relaxation times indicate significant uptake of CSG-IO-NP irrespective of tumour size, whereas the uptake of CREKA-IO-NP is only consistent in small tumours of less than 3 mm in diameter. Larger tumours with significantly reduced tumour blood vessels show a lack of CREKA-IO-NP uptake. Our data suggest CSG-IO-NP are particularly useful for detecting stroma in early and advanced solid tumours.

15.
Nanomedicine ; 5(1): 73-82, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18829396

ABSTRACT

We have used tumor-homing peptides to target abraxane, a clinically approved paclitaxel-albumin nanoparticle, to tumors in mice. The targeting was accomplished with two peptides, CREKA and LyP-1 (CGNKRTRGC). Fluorescein (FAM)-labeled CREKA-abraxane, when injected intravenously into mice bearing MDA-MB-435 human cancer xenografts, accumulated in tumor blood vessels, forming aggregates that contained red blood cells and fibrin. FAM-LyP-1-abraxane co-localized with extravascular islands expressing its receptor, p32. Self-assembled mixed micelles carrying the homing peptide and the label on different subunits accumulated in the same areas of tumors as LyP-1-abraxane, showing that Lyp-1 can deliver intact nanoparticles into extravascular sites. Untargeted, FAM-abraxane was detected in the form of a faint meshwork in tumor interstitium. LyP-1-abraxane produced a statistically highly significant inhibition of tumor growth compared with untargeted abraxane. These results show that nanoparticles can be effectively targeted into extravascular tumor tissue and that targeting can enhance the activity of a therapeutic nanoparticle.


Subject(s)
Breast Neoplasms/drug therapy , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Albumin-Bound Paclitaxel , Albumins/administration & dosage , Albumins/therapeutic use , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Humans , Immunohistochemistry , Mice , Mice, Nude , Peptides/administration & dosage , Peptides/chemistry
16.
Biomaterials ; 219: 119373, 2019 10.
Article in English | MEDLINE | ID: mdl-31374479

ABSTRACT

Oncofetal fibronectin (FN-EDB) and tenascin-C C domain (TNC-C) are nearly absent in extracellular matrix of normal adult tissues but upregulated in malignant tissues. Both FN-EDB and TNC-C are developed as targets of antibody-based therapies. Here we used peptide phage biopanning to identify a novel targeting peptide (PL1, sequence: PPRRGLIKLKTS) that interacts with both FN-EDB and TNC-C. Systemic PL1-functionalized model nanoscale payloads [iron oxide nanoworms (NWs) and metallic silver nanoparticles] homed to glioblastoma (GBM) and prostate carcinoma xenografts, and to non-malignant angiogenic neovessels induced by VEGF-overexpression. Antibody blockage experiments demonstrated that PL1 tumor homing involved interactions with both receptor proteins. Treatment of GBM mice with PL1-targeted model therapeutic nanocarrier (NWs loaded with a proapoptotic peptide) resulted in reduced tumor growth and increased survival, whereas treatment with untargeted particles had no effect. PL1 peptide may have applications as an affinity ligand for delivery of diagnostic and therapeutic compounds to microenvironment of solid tumors.


Subject(s)
Drug Delivery Systems , Fibronectins/metabolism , Neoplasms/drug therapy , Peptides/therapeutic use , Tenascin/metabolism , Amino Acid Sequence , Animals , Apoptosis , Cell Line, Tumor , Ferric Compounds/chemistry , Glioblastoma/drug therapy , Humans , Magnetic Resonance Imaging , Metal Nanoparticles/chemistry , Mice, Nude , Nanoparticles/chemistry , Neoplasms/diagnostic imaging , Silver/chemistry
17.
J Control Release ; 308: 109-118, 2019 08 28.
Article in English | MEDLINE | ID: mdl-31255690

ABSTRACT

Tumor-selective drug conjugates can potentially improve the prognosis for patients affected by glioblastoma (GBM) - the most common and malignant type of brain cancer with no effective cure. Here we evaluated a novel tumor penetrating peptide that targets cell surface p32, LinTT1 (AKRGARSTA), as a GBM targeting ligand for systemically-administered nanoparticles. LinTT1-functionalization increased tumor homing of iron oxide nanoworms (NWs) across a panel of five GBM models ranging from infiltratively-disseminating to angiogenic phenotypes. LinTT1-NWs homed to CD31-positive tumor blood vessels, including to transdifferentiated endothelial cells, and showed co-localization with tumor macrophages and lymphatic vessels. LinTT1 functionalization also resulted in increased GBM delivery of other types of systemically-administered nanoparticles: silver nanoparticles and albumin-paclitaxel nanoparticles. Finally, LinTT1-guided proapoptotic NWs exerted strong anti-glioma activity in two models of GBM, including doubling the lifespan of the mice in an aggressive orthotopic stem cell-like GBM that recapitulates the histological hallmarks of human GBM. Our study suggests that LinTT1 targeting strategy can be used to increase GBM uptake of systemic nanoparticles for improved imaging and therapy.


Subject(s)
Brain Neoplasms/drug therapy , Drug Delivery Systems , Glioblastoma/drug therapy , Nanoparticles , Peptides/administration & dosage , Albumins/administration & dosage , Albumins/pharmacokinetics , Animals , Cell Line, Tumor , Endothelial Cells/metabolism , Female , Ferric Compounds/chemistry , Glioblastoma/pathology , Humans , Male , Metal Nanoparticles , Mice , Mice, Inbred C57BL , Mice, Nude , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Peptides/chemistry , Silver/chemistry , Xenograft Model Antitumor Assays
18.
EMBO Mol Med ; 11(12): e10923, 2019 12.
Article in English | MEDLINE | ID: mdl-31709774

ABSTRACT

High extracellular matrix (ECM) content in solid cancers impairs tumour perfusion and thus access of imaging and therapeutic agents. We have devised a new approach to degrade tumour ECM, which improves uptake of circulating compounds. We target the immune-modulating cytokine, tumour necrosis factor alpha (TNFα), to tumours using a newly discovered peptide ligand referred to as CSG. This peptide binds to laminin-nidogen complexes in the ECM of mouse and human carcinomas with little or no peptide detected in normal tissues, and it selectively delivers a recombinant TNFα-CSG fusion protein to tumour ECM in tumour-bearing mice. Intravenously injected TNFα-CSG triggered robust immune cell infiltration in mouse tumours, particularly in the ECM-rich zones. The immune cell influx was accompanied by extensive ECM degradation, reduction in tumour stiffness, dilation of tumour blood vessels, improved perfusion and greater intratumoral uptake of the contrast agents gadoteridol and iron oxide nanoparticles. Suppressed tumour growth and prolonged survival of tumour-bearing mice were observed. These effects were attainable without the usually severe toxic side effects of TNFα.


Subject(s)
Extracellular Matrix/metabolism , Animals , Cell Line , Cell Surface Display Techniques , Contrast Media/metabolism , Female , Ferric Compounds/metabolism , Gadolinium/metabolism , Heterocyclic Compounds/metabolism , Humans , Male , Mice , Nanoparticles/metabolism , Organometallic Compounds/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Bioconjug Chem ; 19(8): 1570-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18611045

ABSTRACT

The in vivo fate of nanomaterials strongly determines their biomedical efficacy. Accordingly, much effort has been invested into the development of library screening methods to select targeting ligands for a diversity of sites in vivo. Still, broad application of chemical and biological screens to the in vivo targeting of nanomaterials requires ligand attachment chemistries that are generalizable, efficient, covalent, orthogonal to diverse biochemical libraries, applicable under aqueous conditions, and stable in in vivo environments. To date, the copper(I)-catalyzed Huisgen 1,3-dipolar cycloaddition or "click" reaction has shown considerable promise as a method for developing targeted nanomaterials in vitro. Here, we investigate the utility of "click" chemistry for the in vivo targeting of inorganic nanoparticles to tumors. We find that "click" chemistry allows cyclic LyP-1 targeting peptides to be specifically linked to azido-nanoparticles and to direct their binding to p32-expressing tumor cells in vitro. Moreover, "click" nanoparticles are able to stably circulate for hours in vivo following intravenous administration (>5 h circulation time), extravasate into tumors, and penetrate the tumor interstitium to specifically bind p32-expressing cells in tumors. In the future, in vivo use of "click" nanomaterials should expedite the progression from ligand discovery to in vivo evaluation and diversify approaches toward multifunctional nanoparticle development.


Subject(s)
Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Peptides, Cyclic/chemistry , Peptides, Cyclic/therapeutic use , Alkynes/chemistry , Amines/chemistry , Amino Acid Sequence , Animals , Azides/chemistry , Carrier Proteins , Cattle , Cell Line, Tumor , Fluorescence , Gene Expression Regulation , Humans , Magnetics , Mice , Mitochondrial Proteins/metabolism , Peptides, Cyclic/metabolism , Polymers/chemistry , Sensitivity and Specificity , Sulfhydryl Compounds/chemistry
20.
Biomaterials ; 166: 52-63, 2018 06.
Article in English | MEDLINE | ID: mdl-29544111

ABSTRACT

Cationic liposome-nucleic acid (CL-NA) complexes, which form spontaneously, are a highly modular gene delivery system. These complexes can be sterically stabilized via PEGylation [PEG: poly (ethylene glycol)] into nanoparticles (NPs) and targeted to specific tissues and cell types via the conjugation of an affinity ligand. However, there are currently no guidelines on how to effectively navigate the large space of compositional parameters that modulate the specific and nonspecific binding interactions of peptide-targeted NPs with cells. Such guidelines are desirable to accelerate the optimization of formulations with novel peptides. Using PEG-lipids functionalized with a library of prototypical tumor-homing peptides, we varied the peptide density and other parameters (binding motif, peptide charge, CL/DNA charge ratio) to study their effect on the binding and uptake of the corresponding NPs. We used flow cytometry to quantitatively assess binding as well as internalization of NPs by cultured cancer cells. Surprisingly, full peptide coverage resulted in less binding and internalization than intermediate coverage, with the optimum coverage varying between cell lines. In, addition, our data revealed that great care must be taken to prevent nonspecific electrostatic interactions from interfering with the desired specific binding and internalization. Importantly, such considerations must take into account the charge of the peptide ligand as well as the membrane charge density and the CL/DNA charge ratio. To test our guidelines, we evaluated the in vivo tumor selectivity of selected NP formulations in a mouse model of peritoneally disseminated human gastric cancer. Intraperitoneally administered peptide-tagged CL-DNA NPs showed tumor binding, minimal accumulation in healthy control tissues, and preferential penetration of smaller tumor nodules, a highly clinically relevant target known to drive recurrence of the peritoneal cancer.


Subject(s)
DNA , Gene Transfer Techniques , Liposomes , Nanoparticles , Peptides , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cations , DNA/chemistry , Genetic Therapy/methods , Humans , Lipids/chemistry , Liposomes/chemistry , Nanoparticles/chemistry , Peptides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL