Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
3.
Nat Nanotechnol ; 18(2): 184-192, 2023 02.
Article in English | MEDLINE | ID: mdl-36702954

ABSTRACT

Cancer therapies often have narrow therapeutic indexes and involve potentially suboptimal combinations due to the dissimilar physical properties of drug molecules. Nanomedicine platforms could address these challenges, but it remains unclear whether synergistic free-drug ratios translate to nanocarriers and whether nanocarriers with multiple drugs outperform mixtures of single-drug nanocarriers at the same dose. Here we report a bottlebrush prodrug (BPD) platform designed to answer these questions in the context of multiple myeloma therapy. We show that proteasome inhibitor (bortezomib)-based BPD monotherapy slows tumour progression in vivo and that mixtures of bortezomib, pomalidomide and dexamethasone BPDs exhibit in vitro synergistic, additive or antagonistic patterns distinct from their corresponding free-drug counterparts. BPDs carrying a statistical mixture of three drugs in a synergistic ratio outperform the free-drug combination at the same ratio as well as a mixture of single-drug BPDs in the same ratio. Our results address unanswered questions in the field of nanomedicine, offering design principles for combination nanomedicines and strategies for improving current front-line monotherapies and combination therapies for multiple myeloma.


Subject(s)
Multiple Myeloma , Prodrugs , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Bortezomib/therapeutic use , Dexamethasone/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology
5.
Polym Chem ; 11(29): 4768-4779, 2020 Aug 07.
Article in English | MEDLINE | ID: mdl-33790990

ABSTRACT

Nitroxide-based organic-radical contrast agents (ORCAs) are promising as safe, next-generation magnetic resonance imaging (MRI) tools. Nevertheless, stimuli-responsive ORCAs that enable MRI monitoring of prodrug activation have not been reported; such systems could open new avenues for prodrug validation and image-guided drug delivery. Here, we introduce a novel "pro-ORCA" concept that addresses this challenge. By covalent conjugation of nitroxides and drug molecules (doxorubicin, DOX) to the same brush-arm star polymer (BASP) through chemically identical cleavable linkers, we demonstrate that pro-ORCA and prodrug activation, i.e., ORCA and DOX release, leads to significant changes in MRI contrast that correlate with cytotoxicity. This approach is shown to be general for a range of commonly used linker cleavage mechanisms (e.g., photolysis and hydrolysis) and release rates. Pro-ORCAs could find applications as research tools or clinically viable "reporter theranostics" for in vitro and in vivo MRI-correlated prodrug activation.

6.
Nanoscale ; 11(43): 20485-20496, 2019 Nov 21.
Article in English | MEDLINE | ID: mdl-31650133

ABSTRACT

Monitoring malignant progression and disease recurrence post-therapy are central challenges to improving the outcomes of patients with multiple myeloma (MM). Whereas current detection methods that rely upon bone marrow examination allow for precise monitoring of minimal residual disease and can help to elucidate clonal evolution, they do not take into account the spatial heterogeneity of the tumor microenvironment. As such, they are uninformative as to the localization of malignant plasma cells and may lead to false negative results. With respect to the latter challenge, clinically-available imaging agents are neither sufficiently sensitive nor specific enough to detect minute plasma cell populations. Here, we sought to explore methods by which to improve detection of MM cells within their natural bone marrow environment, using whole-animal magnetic resonance imaging to longitudinally monitor early-stage disease as well as to enhance tumor detection after systemic therapy. We conducted a proof-of-concept study to demonstrate that ultra-small (<5 nm) gadolinium-containing nanoparticles bound to full-length antibodies against the B-cell maturation antigen (BCMA) exhibit rapid tumor uptake followed by renal clearance, improving the signal-to-noise ratio for MM detection beyond levels that are currently afforded by other FDA-approved clinical imaging modalities. We anticipate that when combined with bone marrow or blood biopsy, such imaging constructs could help to augment the effective management of patients with MM.


Subject(s)
Antibodies/chemistry , Multiple Myeloma/diagnosis , Nanoparticles/chemistry , Animals , Antibodies/immunology , Antibodies/metabolism , B-Cell Maturation Antigen/immunology , Bone Marrow/metabolism , Bone Marrow/pathology , Contrast Media/chemistry , Contrast Media/pharmacokinetics , Disease Models, Animal , Early Detection of Cancer , Gadolinium/chemistry , Humans , Magnetic Resonance Imaging , Mice , Mice, SCID , Microscopy, Fluorescence , Multiple Myeloma/pathology , Nanoparticles/metabolism , Neoplasm, Residual/diagnosis , Plasma Cells/metabolism , Plasma Cells/pathology , Signal-To-Noise Ratio , Signaling Lymphocytic Activation Molecule Family/immunology , Tissue Distribution
7.
ACS Nano ; 12(11): 11343-11354, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30387988

ABSTRACT

Nitroxides occupy a privileged position among plausible metal-free magnetic resonance imaging (MRI) contrast agents (CAs) due to their inherently low-toxicity profiles; nevertheless, their translational development has been hindered by a lack of appropriate contrast sensitivity. Nanostructured materials with high nitroxide densities, where each individual nitroxide within a macromolecular construct contributes to the image contrast, could address this limitation, but the synthesis of such materials remains challenging. Here, we report a modular and scalable synthetic approach to nitroxide-based brush-arm star polymer (BASP) organic radical CAs (ORCAs) with high nitroxide loadings. The optimized ∼30 nm diameter "BASP-ORCA3" displays outstanding T2 sensitivity with a very high molecular transverse relaxivity ( r2 > 1000 mM-1 s-1). BASP-ORCA3 further exhibits excellent stability in vivo, no acute toxicity, and highly desirable pharmacokinetic and biodistribution profiles for longitudinal detection of tumors by MRI. When injected intravenously into mice bearing subcutaneous plasmacytomas, BASP-ORCA3 affords distinct in vivo visualization of tumors on translationally relevant time scales. Leveraging its high sensitivity, BASP-ORCA3 enables efficient mapping of tumor necrosis, which is an important biomarker to predict therapeutic outcomes. Moreover, BASP-ORCA3 allows for detection of millimetric tumor implants in a disseminated murine model of advanced-stage human ovarian cancer that possess genetic, histological, and vascular characteristics that are similar to those seen in patients. This work establishes BASP-ORCA3 as a promising metal-free spin contrast agent for MRI.


Subject(s)
Magnetic Resonance Imaging , Neoplasms, Experimental/diagnostic imaging , Nitrogen Oxides/chemistry , Optical Imaging , Polymers/chemistry , Animals , Humans , Mice , Mice, Inbred BALB C , Molecular Structure
8.
JCI Insight ; 3(12)2018 06 21.
Article in English | MEDLINE | ID: mdl-29925694

ABSTRACT

Mucin 1 (MUC1) is a heterodimeric protein that is aberrantly overexpressed on the surface of diverse human carcinomas and is an attractive target for the development of mAb-based therapeutics. However, attempts at targeting the shed MUC1 N-terminal subunit have been unsuccessful. We report here the generation of mAb 3D1 against the nonshed oncogenic MUC1 C-terminal (MUC1-C) subunit. We show that mAb 3D1 binds with low nM affinity to the MUC1-C extracellular domain at the restricted α3 helix. mAb 3D1 reactivity is selective for MUC1-C-expressing human cancer cell lines and primary cancer cells. Internalization of mAb 3D1 into cancer cells further supported the conjugation of mAb 3D1 to monomethyl auristatin E (MMAE). The mAb 3D1-MMAE antibody-drug conjugate (ADC) (a) kills MUC1-C-positive cells in vitro, (b) is nontoxic in MUC1-transgenic (MUC1.Tg) mice, and (c) is active against human HCC827 lung tumor xenografts. Humanized mAb (humAb) 3D1 conjugated to MMAE also exhibited antitumor activity in (a) MUC1.Tg mice harboring syngeneic MC-38/MUC1 tumors, (b) nude mice bearing human ZR-75-1 breast tumors, and (c) NCG mice engrafted with a patient-derived triple-negative breast cancer. These findings and the absence of associated toxicities support clinical development of humAb 3D1-MMAE ADCs as a therapeutic for the many cancers with MUC1-C overexpression.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunoconjugates/pharmacology , Mucin-1/immunology , Oncogene Proteins/immunology , Animals , Antibodies, Monoclonal/chemistry , Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Carcinoma/pathology , Cell Line, Tumor , Female , Humans , Immunoconjugates/chemistry , Immunoconjugates/therapeutic use , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Models, Molecular , Mucin-1/chemistry , Oligopeptides , Oncogene Proteins/chemistry , Protein Conformation, alpha-Helical , Xenograft Model Antitumor Assays
9.
Trends Mol Med ; 24(6): 560-574, 2018 06.
Article in English | MEDLINE | ID: mdl-29773319

ABSTRACT

In the past decades, considerable progress has been made in our understanding and treatment of multiple myeloma. Several challenges remain including our abilities to longitudinally image tumor responses to treatment, to combine various therapeutic agents with different mechanisms of action but with overlapping toxicities, and to efficiently harness the power of the immune system to augment remission and/or to induce permanent cures. Nanomedicine may help to address many of these outstanding issues, affording novel diagnostic capabilities and offering disruptive technologies that promise to revolutionize treatment. Here, we review recent developments and the future of nanomedicine for multiple myeloma, highlighting new considerations in nanoparticle designs that may help to augment active targeting, to facilitate longitudinal imaging, and to improve drug delivery.


Subject(s)
Multiple Myeloma/diagnosis , Multiple Myeloma/therapy , Nanomedicine , Animals , Disease Management , Disease Models, Animal , Humans , Nanomedicine/methods , Theranostic Nanomedicine
10.
Nano Lett ; 18(4): 2195-2208, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29533667

ABSTRACT

Human pancreatic ductal adenocarcinoma (PDAC) contains a distinctively dense stroma that limits the accessibility of anticancer drugs, contributing to its poor overall prognosis. Nanoparticles can enhance drug delivery and retention in pancreatic tumors and have been utilized clinically for their treatment. In preclinical studies, various mouse models differentially recapitulate the microenvironmental features of human PDAC. Here, we demonstrate that through utilization of different organic cosolvents and by doping of a homopolymer of poly(ε-caprolactone), a diblock copolymer composition of poly(ethylene oxide)- block-poly(ε-caprolactone) may be utilized to generate biodegradable and nanoscale micelles with different physical properties. Noninvasive optical imaging was employed to examine the pharmacology and biodistribution of these various nanoparticle formulations in both allografted and autochthonous mouse models of PDAC. In contrast to the results reported with transplanted tumors, spherical micelles as large as 300 nm in diameter were found to extravasate in the autochthonous model, reaching a distance of approximately 20 µm from the nearest tumor cell clusters. A lipophilic platinum(IV) prodrug of oxaliplatin was further able to achieve a ∼7-fold higher peak accumulation and a ∼50-fold increase in its retention half-life in pancreatic tumors when delivered with 100 nm long worm-like micelles as when compared to the free drug formulation of oxaliplatin. Through further engineering of nanoparticle properties, as well as by widespread adoption of the autochthonous tumor model for preclinical testing, future therapeutic formulations may further enhance the targeting and penetration of anticancer agents to improve survival outcomes in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/diagnostic imaging , Lactones/analysis , Nanoparticles/analysis , Neoplasm Transplantation/diagnostic imaging , Neoplasms, Experimental/diagnostic imaging , Pancreatic Neoplasms/diagnostic imaging , Polyethylene Glycols/analysis , Animals , Antineoplastic Agents/administration & dosage , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Female , Humans , Lactones/pharmacokinetics , Mice , Mice, Nude , Micelles , Neoplasms, Experimental/drug therapy , Optical Imaging/methods , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Polyethylene Glycols/pharmacokinetics
11.
Nat Commun ; 9(1): 628, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416025

ABSTRACT

The original version of this Article contained an error in the spelling of the author Yingjie Yu, which was incorrectly given as Yu Yingjie. Furthermore, in Figure 3a, the labels 'MD | p < 0.05' incorrectly read 'MD | p > 0.05'. These errors have now been corrected in both the PDF and HTML versions of the Article.

12.
Nat Commun ; 8(1): 2166, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29255160

ABSTRACT

Advanced-stage epithelial ovarian cancers are amongst the most difficult to treat tumors and have proven to be refractory to most cytotoxic, molecularly targeted, or immunotherapeutic approaches. Here, we report that nanoparticle-drug conjugates (NDCs) of monomethyl auristatin E (MMAE) significantly increase loading on a per-vehicle basis as compared to antibody-drug conjugates (ADCs). Their intraperitoneal administration enabled triggered release of the active MMAE toxin to inhibit tumor growth and to extend animal survival to >90 days in a cell-line xenograft model of disseminated ovarian cancer. In a patient-derived xenograft model of advanced-stage and platinum-resistant ovarian cancer, an MMAE-based NDC doubled the duration of tumor growth inhibition as compared to cisplatin. NDCs of highly potent toxins thus introduce a translatable platform that may be exploited to maximize the safety and efficacy of cytotoxic chemotherapies, combining the best features of ADCs with those of nanoparticle-based therapeutics.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Immunoconjugates/pharmacology , Nanoparticles/chemistry , Ovarian Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , Immunoconjugates/chemistry , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/pathology , Survival Analysis , Treatment Outcome
13.
Nanoscale ; 9(36): 13465-13476, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28861570

ABSTRACT

Motivated by the goal of developing a fully biodegradable optical contrast agent with translational clinical potential, a nanoparticle delivery vehicle was generated from the self-assembly of poly(ethylene-glycol)-block-poly(trimethylene carbonate-co-caprolactone) (PEG-b-TCL) copolymers. Cryogenic transmission electron microscopy verified that PEG-b-TCL-based micelles were formed at low solution temperatures (∼38 °C). Detailed spectroscopic experiments validated facile loading of large quantities of the high emission dipole strength, tris(porphyrin)-based fluorophore PZn3 within their cores, and the micelles displayed negligible in vitro and in vivo toxicities in model systems. The pharmacokinetics and biodistribution of PZn3-loaded PEG-b-TCL-based micelles injected intravenously were determined via ex vivo near-infrared (NIR) imaging of PZn3 emission in microcapillary tubes containing minute quantities of blood, to establish a novel method for minimally invasive pharmacokinetic monitoring. The in vivo circulatory half-life of the PEG-b-TCL-based micelles was found to be ∼19.6 h. Additionally, longitudinal in vivo imaging of orthotopically transplanted breast tumors enabled determination of micelle biodistribution that correlated to ex vivo imaging results, demonstrating accumulation predominantly within the tumors and livers of mice. The PEG-b-TCL-based micelles quickly extravasated within 4T1 orthotopic mammary carcinomas, exhibiting peak accumulation at ∼48 h following intravenous tail-vein injection. In summary, PEG-b-TCL-based micelles demonstrated favorable characteristics for further development as in vivo optical contrast agents for minimally invasive imaging of breast tumors.


Subject(s)
Breast Neoplasms/diagnostic imaging , Contrast Media , Micelles , Polyesters , Polyethylene Glycols , Animals , Female , Human Umbilical Vein Endothelial Cells , Humans , Infrared Rays , Mice , Mice, Inbred BALB C , Tissue Distribution
14.
Biomaterials ; 134: 202-215, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28482280

ABSTRACT

Cell-intrinsic reporters such as luciferase (LUC) and red fluorescent protein (RFP) have been commonly utilized in preclinical studies to image tumor growth and to monitor therapeutic responses. While extrinsic reporters that emit near infrared I (NIR-I: 650-950 nm) or near-infrared II (NIR-II: 1000-1700 nm) optical signals have enabled minimization of tissue autofluorescence and light scattering, it has remained unclear as to whether their use has afforded more accurate tumor imaging in small animals. Here, we developed a novel optical imaging construct comprised of rare earth lanthanide nanoparticles coated with biodegradable diblock copolymers and doped with organic fluorophores, generating NIR-I and NIR-II emissive bands upon optical excitation. Simultaneous injection of multiple spectrally-unique nanoparticles into mice bearing tumor implants established via intraperitoneal dissemination of LUC+/RFP+ OVCAR-8 ovarian cancer cells enabled direct comparisons of imaging with extrinsic vs. intrinsic reporters, NIR-II vs. NIR-I signals, as well as targeted vs. untargeted exogenous contrast agents in the same animal and over time. We discovered that in vivo optical imaging at NIR-II wavelengths facilitates more accurate detection of smaller and earlier tumor deposits, offering enhanced sensitivity, improved spatial contrast, and increased depths of tissue penetration as compared to imaging with visible or NIR-I fluorescent agents. Our work further highlights the hitherto underappreciated enhancements in tumor accumulation that may be achieved with intraperitoneal as opposed to intravenous administration of nanoparticles. Lastly, we found discrepancies in the fidelity of tumor uptake that could be obtained by utilizing small molecules for in vivo as opposed to in vitro targeting of nanoparticles to disseminated tumors.


Subject(s)
Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/diagnosis , Spectroscopy, Near-Infrared/methods , Animals , Cell Line, Tumor , Early Detection of Cancer/methods , Female , Humans , Mice , Mice, Nude , Nanoparticles/chemistry , Xenograft Model Antitumor Assays
15.
J Am Chem Soc ; 139(8): 3033-3044, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28166401

ABSTRACT

RNAi approaches have been widely combined with platinum-based anticancer agents to elucidate cellular responses and to target gene products that mediate acquired resistance. Recent work has demonstrated that platination of siRNA prior to transfection may negatively influence RNAi efficiency based on the position and sequence of its guanosine nucleosides. Here, we used detailed spectroscopic characterization to demonstrate rapid formation of Pt-guanosine adducts within 30 min after coincubation of oxaliplatin [OxaPt(II)] or cisplatin [CisPt(II)] with either guanosine monophosphate or B-cell lymphoma 2 (BCL-2) siRNA. After 3 h of exposure to these platinum(II) agents, >50% of BCL-2 siRNA transcripts were platinated and unable to effectively suppress mRNA levels. Platinum(IV) analogues [OxaPt(IV) or CisPt(IV)] did not form Pt-siRNA adducts but did display decreased in vitro uptake and reduced potency. To overcome these challenges, we utilized biodegradable methoxyl-poly(ethylene glycol)-block-poly(ε-caprolactone)-block-poly(l-lysine) (mPEG-b-PCL-b-PLL) to generate self-assembled micelles that covalently conjugated OxaPt(IV) and/or electrostatically complexed siRNA. We then compared multiple strategies by which to combine BCL-2 siRNA with either OxaPt(II) or OxaPt(IV). Overall, we determined that the concentrations of siRNA (nM) and platinum(II)-based anticancer agents (µM) that are typically used for in vitro experiments led to rapid Pt-siRNA adduct formation and ineffective RNAi. Coincorporation of BCL-2 siRNA and platinum(IV) analogues in a single micelle enabled maximal suppression of BCL-2 mRNA levels (to <10% of baseline), augmented the intracellular levels of platinum (by ∼4×) and the numbers of resultant Pt-DNA adducts (by >5×), increased the cellular fractions that underwent apoptosis (by ∼4×), and enhanced the in vitro antiproliferative activity of the corresponding platinum(II) agent (by 10-100×, depending on the cancer cell line). When combining RNAi and platinum-based anticancer agents, this generalizable strategy may be adopted to maximize synergy during screening or for therapeutic delivery.


Subject(s)
Antineoplastic Agents/pharmacology , Organoplatinum Compounds/pharmacology , RNA Interference , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , MCF-7 Cells , Micelles , Molecular Structure , Organoplatinum Compounds/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
16.
Pharm Res ; 33(3): 573-89, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26508477

ABSTRACT

PURPOSE: To develop a technique that maximizes the encapsulation of functional proteins within neutrally charged, fully PEGylated and nanoscale polymer vesicles (i.e., polymersomes). METHODS: Three conventional vesicle formation methods were utilized for encapsulation of myoglobin (Mb) in polymersomes of varying size, PEG length, and membrane thickness. Mb concentrations were monitored by UV-Vis spectroscopy, inductively coupled plasma optical emission spectroscopy (ICP-OES) and by the bicinchoninic acid (BCA) assay. Suspensions were subject to protease treatment to differentiate the amounts of surface-associated vs. encapsulated Mb. Polymersome sizes and morphologies were monitored by dynamic light scattering (DLS) and by cryogenic transmission electron microscopy (cryo-TEM), respectively. Binding and release of oxygen were measured using a Hemeox analyzer. RESULTS: Using the established "thin-film rehydration" and "direct hydration" methods, Mb was found to be largely surface-associated with negligible aqueous encapsulation within polymersome suspensions. Through iterative optimization, a novel "progressive saturation" technique was developed that greatly increased the final concentrations of Mb (from < 0.5 to > 2.0 mg/mL in solution), the final weight ratio of Mb-to-polymer that could be reproducibly obtained (from < 1 to > 4 w/w% Mb/polymer), as well as the overall efficiency of Mb encapsulation (from < 5 to > 90%). Stable vesicle morphologies were verified by cryo-TEM; the suspensions also displayed no signs of aggregate formation for > 2 weeks as assessed by DLS. "Progressive saturation" was further utilized for the encapsulation of a variety of other proteins, ranging in size from 17 to 450 kDa. CONCLUSIONS: Compared to established vesicle formation methods, "progressive saturation" increases the quantities of functional proteins that may be encapsulated in nanoscale polymersomes.


Subject(s)
Drug Carriers/chemistry , Nanoparticles/chemistry , Polymers/chemistry , Proteins/chemistry , Microscopy, Electron, Transmission/methods , Myoglobin/chemistry , Nanotechnology/methods , Particle Size , Polyethylene Glycols/chemistry , Suspensions/chemistry
17.
Trends Biotechnol ; 32(9): 466-73, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24929580

ABSTRACT

Here, we discuss recent advances in the development of artificial red blood cell (RBC) substitutes, illustrating lessons learned from initial attempts using perfluorocarbon (PFC) emulsions and acellular hemoglobin-based oxygen carriers (HBOCs). We also highlight novel oxygen-containing microparticles, nanoparticles, and stem cell-derived RBC products, with emphasis on improvements in biocompatibility and oxygen delivery. In addition, we envision future developments for the rational design of advanced blood substitutes that aim to address unmet clinical needs.


Subject(s)
Blood Substitutes/pharmacology , Blood Substitutes/therapeutic use , Nanoparticles/therapeutic use , Oxygen/metabolism , Stem Cells/physiology , Biomedical Research/trends , Fluorocarbons/metabolism , Humans
18.
Nanoscale ; 5(22): 10908-15, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24056924

ABSTRACT

Nanoparticles formed from diblock copolymers of FDA approved PEO and PCL have generated considerable interest as in vivo drug delivery vehicles. Herein, we report the synthesis of the most extensive family PEO-b-PCL copolymers that vary over the largest range of number-average molecular weights (Mn: 3.6-57k), PEO weight fractions (fPEO: 0.08-0.33), and PEO chain lengths (0.75-5.8k) reported to date. These polymers were synthesized in order to establish the full range of aqueous phase behaviours of these diblock copolymers and to specifically identify formulations that were able to generate bilayered vesicles (polymersomes). Cryogenic transmission electron microscopy (cryo-TEM) was utilized in order to visualize the morphology of these structures upon aqueous self-assembly of dry polymer films. Nanoscale polymersomes were formed from PEO-b-PCL copolymers over a wide range of PEO weight fractions (fPEO: 0.14-0.27) and PEO molecular weights (0.75-3.8k) after extrusion of aqueous suspensions. Comparative morphology diagrams, which describe the nature of self-assembled structures as a function of diblock copolymer molecular weight and PEO weight fraction, show that in contrast to micron-scale polymersomes, which form only from a limited range of PEO-b-PCL diblock copolymer compositions, a multiplicity of PEO-b-PCL diblock copolymer compositions are able to give rise to nanoscale vesicles. These data underscore that PEO-b-PCL compositions that spontaneously form micron-sized polymersomes, as well as those that have previously been reported to form polymersomes via a cosolvent fabrication system, provide only limited insights into the distribution of PEO-b-PCL diblocks that give rise to nanoscale vesicles. The broad range of polymersome-forming PEO-b-PCL compositions described herein suggest the ability to construct extensive families of nanoscale vesicles of varied bilayer thickness, providing the ability to tune the timescales of vesicle degradation and encapsulant release based on the intended in vivo application.


Subject(s)
Drug Carriers/chemistry , Lipid Bilayers/chemistry , Nanostructures/chemistry , Polyesters/chemistry , Lipid Bilayers/metabolism , Polyesters/chemical synthesis , Water/chemistry
19.
Article in English | MEDLINE | ID: mdl-20049787

ABSTRACT

In vivo fluorescence imaging with near-infrared (NIR) light holds enormous potential for a wide variety of molecular diagnostic and therapeutic applications. Because of its quantitative sensitivity, inherent biological safety, and relative ease of use (i.e., with respect to cost, time, mobility, and its familiarity to a diverse population of investigators), fluorescence-based imaging techniques are being increasingly utilized in small-animal research. Moreover, there is substantial interest in the translation of novel optical techniques into the clinic, where they will prospectively aid in noninvasive and quantitative screening, disease diagnosis, and post-treatment monitoring of patients. Effective deep-tissue fluorescence imaging requires the application of exogenous NIR-emissive contrast agents. Currently, available probes fall into two major categories: organic and inorganic NIR fluorophores (NIRFs). In the studies reviewed herein, we utilized polymersomes (50 nm to 50 microm diameter polymer vesicles) for the incorporation and delivery of large numbers of highly emissive oligo (porphyrin)-based, organic NIRFs.


Subject(s)
Contrast Media/chemistry , Drug Carriers/chemical synthesis , Fluorescent Dyes/chemistry , Microscopy, Fluorescence/methods , Nanoparticles/chemistry , Molecular Probe Techniques , Nanoparticles/ultrastructure
20.
Faraday Discuss ; 139: 129-41; discussion 213-28, 419-20, 2008.
Article in English | MEDLINE | ID: mdl-19048993

ABSTRACT

Polymersomes are vesicles whose membranes are comprised of self-assembled amphiphilic block co-polymers. Synthetic control of block co-polymer chemistry provides an advantageous diversity of polymersome functions, ranging from tunable materials strength, superior encaspulation of hydrophobic and hydrophilic drugs and optical dyes, and facile functionalization. We have exploited polymersome tunability to make leuko-polymersomes: polymersomes with the adhesive properties of leukocytes. By functionalizing the terminal groups on the outer shell of the vesicle with biotin, we have used modular avidin-biotin chemistry to attach adhesion ligands that mimic the two critical adhesion pathways that leukocytes utilize to achieve adhesion in the fast fluid flow of blood vessels--selectins and integrins. We demonstrate that adhesion is specific and is supported at hydrodynamic flow rates at which leukocytes adhere. We envision the use of such particles for monitoring or treating inflammation, cancer and cardiovascular disease.


Subject(s)
Leukocytes/physiology , Polymers/chemistry , Cell Adhesion
SELECTION OF CITATIONS
SEARCH DETAIL
...