Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Control Release ; 354: 244-259, 2023 02.
Article in English | MEDLINE | ID: mdl-36596340

ABSTRACT

Nanoparticle (NP) technology holds significant promise to mediate targeted drug delivery to specific organs in the body. Understanding the 3D biodistribution of NPs in heterogeneous environments such as the tumor tissue can provide crucial information on efficacy, safety and potential clinical outcomes. Here we present a novel end-to-end workflow, VIOLA, which makes use of tissue clearing methodology in conjunction with high resolution imaging and advanced 3D image processing to quantify the spatiotemporal 3D biodistribution of fluorescently labeled ACCURIN® NPs. Specifically, we investigate the spatiotemporal biodistribution of NPs in three different murine tumor models (CT26, EMT6, and KPC-GEM) of increasing complexity and translational relevance. We have developed new endpoints to characterize NP biodistribution at multiple length scales. Our observations reveal that the macroscale NP biodistribution is spatially heterogeneous and exhibits a gradient with relatively high accumulation at the tumor periphery that progressively decreases towards the tumor core in all the tumor models. Microscale analysis revealed that NP extravasation from blood vessels increases in a time dependent manner and plateaus at 72 h post injection. Volumetric analysis and pharmacokinetic modeling of NP biodistribution in the vicinity of the blood vessels revealed that the local NP density exhibits a distance dependent spatiotemporal biodistribution which provide insights into the dynamics of NP extravasation in the tumor tissue. Our data represents a comprehensive analysis of NP biodistribution at multiple length scales in different tumor models providing unique insights into their spatiotemporal dynamics. Specifically, our results show that NPs exhibit a dynamic equilibrium with macroscale heterogeneity combined with microscale homogeneity.


Subject(s)
Nanoparticles , Neoplasms , Viola , Animals , Mice , Tissue Distribution , Drug Delivery Systems
2.
J Med Chem ; 62(21): 9541-9559, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31593466

ABSTRACT

Novel nanoparticle-drug conjugates (NDCs) containing diverse, clinically relevant anticancer drug payloads (docetaxel, cabazitaxel, and gemcitabine) were successfully generated and tested in drug discovery studies. The NDCs utilized structurally varied linkers that attached the drug payloads to a ß-cyclodextrin-PEG copolymer to form self-assembled nanoparticles. In vitro release studies revealed a diversity of release rates driven by linker structure-activity relationships (SARs). Improved in vivo pharmacokinetics (PK) for the cabazitaxel (CBTX) NDCs with glycinate-containing (1c) and hexanoate-containing linkers (2c) were demonstrated, along with high and sustained tumor levels (>168 h of released drug in tumor tissues). This led to potent efficacy and survival in both taxane- and docetaxel-resistant in vivo anticancer mouse efficacy models. Overall, the CBTX-hexanoate NDC 2c (CRLX522), demonstrated optimal and improved in vivo PK (plasma and tumor) and efficacy profile versus those of the parent drug, and the results support the potential therapeutic use of CRLX522 as a new anticancer agent.


Subject(s)
Drug Carriers/chemistry , Drug Design , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Taxoids/chemistry , Taxoids/pharmacology , beta-Cyclodextrins/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Male , Melanoma, Experimental/pathology , Mice , Taxoids/pharmacokinetics , Tissue Distribution
3.
Bioorg Chem ; 92: 103233, 2019 11.
Article in English | MEDLINE | ID: mdl-31518759

ABSTRACT

Broussonetia papyrifera has been used as a diuretic, tonic and suppressor of edema. Bioactivity-guided fractionation and metabolite investigation of root bark extracts of this plant resulted in the isolation and identification of six 1,3-diphenylpropanes (1, 2, 8, 10, 17, 20), flavanone (3), two chalcones (4, 5), five flavans (6, 11, 14-16), dihydroflavonol (7) and five flavonols (9, 12, 13, 18, 19), including five new compounds (5, 7, 8, 19, 20) that inhibit NO production in LPS-induced RAW264.7 cells. The structures of compounds 1-20 were elucidated on the basis of spectroscopic data (1D and 2D NMR, MS, MS/MS, and HRMS). In particular, compounds 3, 5, 7, 12, and 20 exhibited significant inhibitory effects on the NO, iNOS, and pro-inflammatory cytokine (TNF-α and IL-6) production. Therefore, this study suggests that the flavonoid-rich products of B. papyrifera, including the new compounds, could be valuable candidates for the development of pharmaceuticals or functional foods in the prevention and treatment of anti-inflammatory disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Broussonetia/chemistry , Flavonoids/pharmacology , Plant Bark/chemistry , Animals , Anti-Inflammatory Agents/isolation & purification , Cell Survival/drug effects , Cyclooxygenase 2/genetics , Dose-Response Relationship, Drug , Flavonoids/isolation & purification , Gene Expression/drug effects , Interleukin-6/antagonists & inhibitors , Lipopolysaccharides , Medicine, Korean Traditional , Mice , Molecular Structure , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/antagonists & inhibitors
4.
Mol Pharm ; 13(3): 737-47, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26835715

ABSTRACT

Small interfering RNA (siRNA) therapeutics have potential advantages over traditional small molecule drugs such as high specificity and the ability to inhibit otherwise "undruggable" targets. However, siRNAs have short plasma half-lives in vivo, can induce a cytokine response, and show poor cellular uptake. Formulating siRNA into nanoparticles offers two advantages: enhanced siRNA stability against nuclease degradation beyond what chemical modification alone can provide; and improved site-specific delivery that takes advantage of the enhanced permeability and retention (EPR) effect. Existing delivery systems generally suffer from poor delivery to tumors. Here we describe the formation and biological activity of polymeric nanopharmaceuticals (PNPs) based on biocompatible and biodegradable poly(lactic-co-glycolic acid) (PLGA) conjugated to siRNA via an intracellular cleavable disulfide linker (PLGA-siRNA). Additionally, these PNPs contain (1) PLGA conjugated to polyethylene glycol (PEG) for enhanced pharmacokinetics of the nanocarrier; (2) a cation for complexation of siRNA and charge compensation to avoid high negative zeta potential; and (3) neutral poly(vinyl alcohol) (PVA) to stabilize the PNPs and support the PEG shell to prevent particle aggregation and protein adsorption. The biological data demonstrate that these PNPs achieve prolonged circulation, tumor accumulation that is uniform throughout the tumor, and prolonged tumor-specific knockdown. PNPs employed in this study had no effect on body weight, blood cell count, serum chemistry, or cytokine response at doses >10 times the effective dose. PNPs, therefore, constitute a promising solution for achieving durable siRNA delivery and gene silencing in tumors.


Subject(s)
Colonic Neoplasms/therapy , Gene Silencing , Green Fluorescent Proteins/antagonists & inhibitors , Molecular Chaperones/antagonists & inhibitors , Nanoparticles/administration & dosage , Polymers/chemistry , RNA, Small Interfering/genetics , Animals , Chemistry, Pharmaceutical , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Genetic Therapy/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lactic Acid/chemistry , Mice , Mice, Inbred C57BL , Mice, Nude , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Proc Natl Acad Sci U S A ; 110(37): 15127-32, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23980155

ABSTRACT

Nanoparticles are currently being investigated in a number of human clinical trials. As information on how nanoparticles function in humans is difficult to obtain, animal studies that can be correlative to human behavior are needed to provide guidance for human clinical trials. Here, we report correlative studies on animals and humans for CRLX101, a 20- to 30-nm-diameter, multifunctional, polymeric nanoparticle containing camptothecin (CPT). CRLX101 is currently in phase 2 clinical trials, and human data from several of the clinical investigations are compared with results from multispecies animal studies. The pharmacokinetics of polymer-conjugated CPT (indicative of the CRLX101 nanoparticles) in mice, rats, dogs, and humans reveal that the area under the curve scales linearly with milligrams of CPT per square meter for all species. Plasma concentrations of unconjugated CPT released from CRLX101 in animals and humans are consistent with each other after accounting for differences in serum albumin binding of CPT. Urinary excretion of polymer-conjugated CPT occurs primarily within the initial 24 h after dosing in animals and humans. The urinary excretion dynamics of polymer-conjugated and unconjugated CPT appear similar between animals and humans. CRLX101 accumulates into solid tumors and releases CPT over a period of several days to give inhibition of its target in animal xenograft models of cancer and in the tumors of humans. Taken in total, the evidence provided from animal models on the CRLX101 mechanism of action suggests that the behavior of CRLX101 in animals is translatable to humans.


Subject(s)
Camptothecin/administration & dosage , Cyclodextrins/administration & dosage , Nanoconjugates/administration & dosage , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Area Under Curve , Camptothecin/pharmacokinetics , Camptothecin/therapeutic use , Cell Line, Tumor , Clinical Trials as Topic , Cyclodextrins/pharmacokinetics , Cyclodextrins/therapeutic use , Dogs , Drug Delivery Systems , Female , Humans , Mice , Mice, Nude , Nanoconjugates/chemistry , Nanoconjugates/therapeutic use , Rats , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Translational Research, Biomedical
6.
Invest New Drugs ; 31(4): 986-1000, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23397498

ABSTRACT

Patients with advanced solid malignancies were enrolled to an open-label, single-arm, dose-escalation study, in which CRLX101 was administered intravenously over 60 min among two dosing schedules, initially weekly at 6, 12, and 18 mg/m(2) and later bi-weekly at 12, 15, and 18 mg/m(2). The maximum tolerated dose (MTD) was determined at 15 mg/m(2) bi-weekly, and an expansion phase 2a study was completed. Patient samples were obtained for pharmacokinetic (PK) and pharmacodynamic (PD) assessments. Response was evaluated per RECIST criteria v1.0 every 8 weeks. Sixty-two patients (31 male; median age 63 years, range 39-79) received treatment. Bi-weekly dosing was generally well tolerated with myelosuppression being the dose-limiting toxicity. Among all phase 1/2a patients receiving the MTD (n = 44), most common grade 3/4 adverse events were neutropenia and fatigue. Evidence of systemic plasma exposure to both the polymer-conjugated and unconjugated CPT was observed in all treated patients. Mean elimination unconjugated CPT Tmax values ranged from 17.7 to 24.5 h, and maximum plasma concentrations and areas under the curve were generally proportional to dose for both polymer-conjugated and unconjugated CPT. Best overall response was stable disease in 28 patients (64 %) treated at the MTD and 16 (73 %) of a subset of NSCLC patients. Median progression-free survival (PFS) for patients treated at the MTD was 3.7 months and for the subset of NSCLC patients was 4.4 months. These combined phase 1/2a data demonstrate encouraging safety, pharmacokinetic, and efficacy results. Multinational phase 2 clinical development of CRLX101 across multiple tumor types is ongoing.


Subject(s)
Camptothecin/therapeutic use , Cellulose/therapeutic use , Cyclodextrins/therapeutic use , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Adult , Aged , Area Under Curve , Biopsy , Camptothecin/adverse effects , Camptothecin/blood , Camptothecin/pharmacokinetics , Cellulose/adverse effects , Cellulose/blood , Cellulose/pharmacokinetics , Cyclodextrins/adverse effects , Cyclodextrins/blood , Cyclodextrins/pharmacokinetics , Demography , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Humans , Immunohistochemistry , Male , Maximum Tolerated Dose , Middle Aged , Nanoparticles/adverse effects , Neoplasm Staging , Neoplasms/diagnostic imaging , Tomography, X-Ray Computed , Treatment Outcome
7.
Curr Bioact Compd ; 7(1): 8-14, 2011 Mar.
Article in English | MEDLINE | ID: mdl-22081768

ABSTRACT

CRLX101 (formerly IT-101) is a first-in-class nanopharmaceutical, currently in Phase 2a development, which has been developed by covalently conjugating camptothecin (CPT) to a linear, cyclodextrin-polyethylene glycol (CD-PEG) co-polymer that self-assembles into nanoparticles. As a nanometer-scale drug carrier system, the cyclodextrin polymeric nanoparticle technology, referred to as "CDP", has unique design features and capabilities. Specifically, CRLX101 preclinical and clinical data confirm that CDP can address not only solubility, formulation, toxicity, and pharmacokinetic challenges associated with administration of CPT, but more importantly, can impart unique biological properties that enhance CPT pharmacodynamics and efficacy.

8.
J Control Release ; 153(1): 49-55, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21406204

ABSTRACT

Camptothecin (CPT) is a potent broad-spectrum anticancer agent that acts through inhibition of topoisomerase 1. Clinical development of CPT was unsuccessful due to poor drug solubility, insufficient in vivo stability of the active form, and toxicity. In order to address these issues, a polymeric nanoparticle comprised of cyclodextrin-poly(ethylene glycol) copolymer (CDP) conjugated to CPT (CRLX101) has been developed and Phase 2 clinical studies are ongoing. Camptothecin is conjugated to the polymer in its active form at 10-12 wt.% loading. CRLX101 self-assembles in solution into nanoparticles with an apparent solubility increase of >1000-fold as compared to the parent drug camptothecin. Preclinical studies exhibited CRLX101 pharmacokinetics superior to the parent drug. Drug concentration in tumor relative to plasma and other major organs is consistent with the enhanced permeation and retention (EPR) anticipated from a nanoparticle. Significant anti-tumor activity was observed that is superior when compared to irinotecan across a broad range of xenograft models. Pharmacokinetic data are consistent with the prolonged half-life and increased AUC. The CRLX101 preclinical and clinical data confirm that CDP can address not only solubility, formulation, toxicity, and pharmacokinetic challenges associated with administration of CPT, but more importantly, can impart unique biological properties, that enhance pharmacodynamics and efficacy of camptothecin.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Camptothecin/administration & dosage , Cyclodextrins/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Polyethylene Glycols/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Camptotheca/chemistry , Camptothecin/chemistry , Camptothecin/pharmacokinetics , Camptothecin/therapeutic use , Cell Line, Tumor , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Mice , Solubility
9.
Nanomedicine ; 6(2): 382-90, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19836468

ABSTRACT

The goal of this study was to evaluate the mechanism of cyclodextrin-based nanoparticle (CDP-NP) uptake into a murine glioma model. Using mixed in vitro culture systems, we demonstrated that CDP-NPs were preferentially taken up by BV2 and N9 microglia (MG) cells compared with GL261 glioma cells. Fluorescent microscopy and flow cytometry analysis of intracranial GL261 gliomas confirmed these findings and demonstrated a predominant CDP-NP uptake by macrophages (MPs) and MG within and around the tumor site. Notably, in mice bearing bilateral intracranial tumor, MG and MPs carrying CDP-NPs were able to migrate to the contralateral tumors. In conclusion, these studies better characterize the cellular distribution of CDP-NPs in intracranial tumors and demonstrate that MPs and MG could potentially be used as nanoparticle drug carriers into malignant brain tumors. FROM THE CLINICAL EDITOR: The goal of this study was to evaluate the mechanism of cyclodextrin-based nanoparticle (CDP-NP) uptake into a murine glioma model. CDP-NP was preferentially taken up microglia (MG) cells as compared to glioma cells. A predominant CDP-NP uptake by macrophages and MG was also shown in and around the tumor site. Macrophages and MG could potentially be used as nanoparticle drug carriers into malignant brain tumors.


Subject(s)
Brain Neoplasms/metabolism , Cyclodextrins/chemistry , Cyclodextrins/pharmacokinetics , Drug Carriers/chemistry , Glioma/metabolism , Macrophages/metabolism , Nanoparticles/chemistry , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Glioma/pathology , Mice , Nanoparticles/therapeutic use
10.
Proc Natl Acad Sci U S A ; 106(27): 11394-9, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19564622

ABSTRACT

IT-101, a cyclodextrin polymer-based nanoparticle containing camptothecin, is in clinical development for the treatment of cancer. Multiorgan pharmacokinetics and accumulation in tumor tissue of IT-101 is investigated by using PET. IT-101 is modified through the attachment of a 1,4,7,10-tetraazacyclododecane-1,4,7-Tris-acetic acid ligand to bind (64)Cu(2+). This modification does not affect the particle size and minimally affects the surface charge of the resulting nanoparticles. PET data from (64)Cu-labeled IT-101 are used to quantify the in vivo biodistribution in mice bearing Neuro2A s.c. tumors. The (64)Cu-labeled IT-101 displays a biphasic plasma elimination. Approximately 8% of the injected dose is rapidly cleared as a low-molecular-weight fraction through the kidneys. The remaining material circulates in plasma with a terminal half-life of 13.3 h. Steadily increasing concentrations, up to 11% injected dose per cm(3), are observed in the tumor over 24 h, higher than any other tissue at that time. A 3-compartment model is used to determine vascular permeability and nanoparticle retention in tumors, and is able to accurately represent the experimental data. The calculated tumor vascular permeability indicates that the majority of nanoparticles stay intact in circulation and do not disassemble into individual polymer strands. A key assumption to modeling the tumor dynamics is that there is a "sink" for the nanoparticles within the tumor. Histological measurements using confocal microscopy show that IT-101 localizes within tumor cells and provides the sink in the tumor for the nanoparticles.


Subject(s)
Camptothecin/pharmacokinetics , Cyclodextrins/pharmacokinetics , Nanoparticles , Neoplasms/pathology , Polymers/pharmacokinetics , Positron-Emission Tomography , Whole Body Imaging , Animals , Cell Line, Tumor , Copper/pharmacokinetics , Fluorescent Antibody Technique , Kidney/metabolism , Mice , Mice, SCID , Models, Biological , Tissue Distribution
11.
Int J Nanomedicine ; 3(3): 359-71, 2008.
Article in English | MEDLINE | ID: mdl-18990945

ABSTRACT

A glycinate derivative of alpha-methylprednisolone (MP) was prepared and conjugated to a linear cyclodextrin polymer (CDP) with a loading of 12.4% w/w. The polymer conjugate (CDP-MP) self-assembled into nanoparticles with a size of 27 nm. Release kinetics of MP from the polymer conjugate showed a half-life (t1/2) of 50 h in phosphate buffer solution (PBS) and 19 h in human plasma. In vitro, the proliferation of human lymphocytes was suppressed to a similar extent but with a delayed effect when CDP-MP was compared with free MP. In vivo, CDP-MP was administered intravenously to mice with collagen-induced arthritis and compared with free MP. CDP-MP was administered weekly for six weeks (0.07, 0.7, and 7 mg/kg/week) and MP was administered daily for six weeks (0.01, 0.1, and 1 mg/kg/day). Body weight changes were minimal in all animals. After 28 days, a significant decrease in arthritis score was observed in animals treated weekly with an intermediate or high dose of CDP-MP. Additionally, dorsoplantar swelling was reduced to baseline in animals treated with CDP-MP at the intermediate and high dose level. Histological evaluation showed a reduction in synovitis, pannus formation and disruption of architecture at the highest dose level of CDP-MP. MP administered daily at equivalent cumulative doses showed minimal efficacy in this model. This study demonstrates that conjugation of MP to a cyclodextrin-polymer may improve its efficacy, leading to lower doses and less frequent administration for a safer and more convenient management of rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Cyclodextrins/administration & dosage , Drug Carriers/chemistry , Lymphocytes/cytology , Lymphocytes/drug effects , Methylprednisolone/administration & dosage , Nanoparticles/chemistry , Animals , Arthritis, Rheumatoid/pathology , Cell Proliferation/drug effects , Cells, Cultured , Cyclodextrins/chemistry , Humans , Methylprednisolone/chemistry , Mice , Nanoparticles/ultrastructure , Polymers/chemistry , Treatment Outcome
12.
Chem Commun (Camb) ; (35): 3631-3, 2007 Sep 21.
Article in English | MEDLINE | ID: mdl-17728876

ABSTRACT

Block copolymers with sequences of differential reactivity were synthesized, and the step-wise and selective derivatization to form a new block copolymer was demonstrated.


Subject(s)
Models, Molecular , Polymethacrylic Acids/chemistry , Polymethacrylic Acids/chemical synthesis , Magnetic Resonance Spectroscopy , Molecular Structure
13.
J Control Release ; 122(3): 279-86, 2007 Oct 08.
Article in English | MEDLINE | ID: mdl-17599628

ABSTRACT

Polymers with reactive side chains and narrow molecular weight distributions were synthesized by reversible addition-fragmentation chain transfer (RAFT) polymerization, and the potential to utilize these polymers to prepare drug carriers was demonstrated. p-Nitrophenyl methacrylate (NPMA) and diethoxypropyl methacrylate (DEPMA) were polymerized utilizing cumyl dithiobenzoate (CDB) as the chain transfer agent and azobisisobutyronitrile (AIBN) as the initiator to high conversions (> or = 86%). The resulting pNPMA and pDEPMA had narrow molecular weight distributions (polydispersity indices < 1.3). The ability to functionalize these polymers was confirmed. For pNPMA, up to 86% of the side chains were substituted with the amino acid, glycine methyl ester. The side chains of pDEPMA were hydrolyzed to aldehydes and reaction with O-benzylhydroxylamine and O-methylhydroxylamine to form stable oxime bond conjugates was demonstrated. The percent substitution depended on the feed ratios. Conjugation of an aminooxy-functionalized RGD peptide was also demonstrated.


Subject(s)
Aldehydes/chemistry , Drug Carriers , Esters/chemistry , Polymers , Drug Carriers/chemical synthesis , Drug Carriers/chemistry , Molecular Structure , Molecular Weight , Polymers/chemical synthesis , Polymers/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...