Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Methods Mol Biol ; 2789: 3-17, 2024.
Article in English | MEDLINE | ID: mdl-38506986

ABSTRACT

Nanotechnology for drug delivery has made significant advancements over the last two decades. Innovations have been made in cancer research and development, including chemotherapies, imaging agents, and vaccine strategies, as well as other therapeutic areas, e.g., the recent commercialization of mRNA lipid nanoparticles as vaccines against the SARS-CoV-2 virus. The field has also seen technological advancements to aid in addressing the complex questions posed by these novel therapies. In this latest edition of protocols and methods for nanoparticle characterization, we highlight both old and new methodologies for defining physicochemical properties, present both in vitro and in vivo methods to test for a variety of immunotoxicities, and describe assays used for pharmacological studies to assess drug release and tissue distribution.


Subject(s)
Nanoparticles , Vaccines , Nanomedicine/methods , Nanotechnology/methods , Drug Delivery Systems/methods , Nanoparticles/chemistry
2.
Methods Mol Biol ; 2789: 301-311, 2024.
Article in English | MEDLINE | ID: mdl-38507012

ABSTRACT

The evaluation of temperature-dependent drug release for solubilizing nanoformulations uses a modification of the stable isotope ultrafiltration assay (SITUA). This method is specific to parenterally administered solubilizing nanomedicines and can be used to assess drug release from the total dosage form for regulatory purposes of lot release. The principle upon which this method is based is the relationship between drug solubility and temperature in a plasma simulating media, 4.5% human serum albumin, that allows for discrimination of passing and failing lots based upon the release characteristics.


Subject(s)
Isotopes , Ultrafiltration , Humans , Drug Liberation , Temperature , Solubility
3.
Methods Mol Biol ; 2789: 313-322, 2024.
Article in English | MEDLINE | ID: mdl-38507013

ABSTRACT

A primary issue with nanomedicine biological evaluation is determination of nanoparticle carrier tissue distribution and stability. Here we present a method to evaluate nanomedicine distribution in tissues that is applicable to most nanomedicine constructs. This method utilizes immunohistochemical (IHC) analysis of an Alexa Fluor 488-tag and/or polyethylene glycol (PEG), a very common nanomedicine component, for tissue localization. Using specific Alexa Fluor 488- and/or PEG antibody-based IHC staining procedures allows evaluation of high-resolution nanoparticle tissue distribution, nanoparticle tissue stability, and also allows correlation of distribution with morphological changes. This protocol outlines the methods to follow to ensure proper tissue collection and optimized immunohistochemical staining of Alexa Fluor 488-tag and PEG in tissues.


Subject(s)
Fluoresceins , Fluorescent Dyes , Polyethylene Glycols , Sulfonic Acids , Immunohistochemistry , Nanomedicine , Tissue Distribution
4.
AAPS J ; 25(3): 39, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37041376

ABSTRACT

Paclitaxel (PTX) is a frequently prescribed chemotherapy drug used to treat a wide variety of solid tumors. Oligo(lactic acid)8-PTX prodrug (o(LA)8-PTX) loaded poly(ethylene glycol)-b-poly(lactic acid) (PEG-b-PLA) micelles have higher loading, slower release and higher antitumor efficacy in murine tumor models over PTX-loaded PEG-b-PLA micelles. The goal of this work is to study plasma stability of o(LA)8-PTX-loaded PEG-b-PLA micelles and its pharmacokinetics after IV injection in rats. In rat plasma, o(LA)8-PTX prodrug is metabolized into o(LA)1-PTX and PTX. In human plasma, o(LA)8-PTX is metabolized more slowly into o(LA)2-PTX, o(LA)1-PTX, and PTX. After IV injection of 10 mg/kg PTX-equiv of o(LA)8-PTX prodrug loaded PEG-b-PLA micelles in Sprague-Dawley rats, metabolite abundance in plasma follows the order: o(LA)1-PTX > o(LA)2-PTX > o(LA)4-PTX > o(LA)6-PTX. Bile metabolite profiles of the o(LA)8-PTX prodrug is similar to plasma metabolite profiles. In comparison to equivalent doses of Abraxane®, plasma PTX exposure is two orders of magnitude higher for Abraxane® than PTX from o(LA)8-PTX prodrug loaded PEG-b-PLA micelles, and plasma o(LA)1-PTX exposure is fivefold higher than PTX from Abraxane®, demonstrating heightened plasma metabolite exposure for enhanced antitumor efficacy.


Subject(s)
Paclitaxel , Prodrugs , Rats , Mice , Humans , Animals , Paclitaxel/pharmacokinetics , Lactic Acid , Micelles , Albumin-Bound Paclitaxel , Drug Carriers/pharmacokinetics , Cell Line, Tumor , Rats, Sprague-Dawley , Polymers , Polyesters
5.
Int J Mol Sci ; 24(1)2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36614194

ABSTRACT

Survival from pancreatic cancer is poor because most cancers are diagnosed in the late stages and there are no therapies to prevent the progression of precancerous pancreatic intraepithelial neoplasms (PanINs). Inhibiting mutant KRASG12D, the primary driver mutation in most human pancreatic cancers, has been challenging. The cholecystokinin-B receptor (CCK-BR) is absent in the normal pancreas but becomes expressed in high grade PanIN lesions and is over-expressed in pancreatic cancer making it a prime target for therapy. We developed a biodegradable nanoparticle polyplex (NP) that binds selectively to the CCK-BR on PanINs and pancreatic cancer to deliver gene therapy. PanIN progression was halted and the pancreas extracellular matrix rendered less carcinogenic in P48-Cre/LSL-KrasG12D/+ mice treated with the CCK-BR targeted NP loaded with siRNA to mutant Kras. The targeted NP also slowed proliferation, decreased metastases and improved survival in mice bearing large orthotopic pancreatic tumors. Safety and toxicity studies were performed in immune competent mice after short or long-term exposure and showed no off-target toxicity by histological or biochemical evaluation. Precision therapy with target-specific NPs provides a novel approach to slow progression of advanced pancreatic cancer and also prevents the development of pancreatic cancer in high-risk subjects without toxicity to other tissues.


Subject(s)
Carcinoma in Situ , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Mice , Humans , Animals , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Disease Models, Animal , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/prevention & control , Pancreas/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma in Situ/genetics , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms
6.
Adv Drug Deliv Rev ; 191: 114591, 2022 12.
Article in English | MEDLINE | ID: mdl-36332724

ABSTRACT

Cancer nanotechnologies possess immense potential as therapeutic and diagnostic treatment modalities and have undergone significant and rapid advancement in recent years. With this emergence, the complexities of data standards in the field are on the rise. Data sharing and reanalysis is essential to more fully utilize this complex, interdisciplinary information to answer research questions, promote the technologies, optimize use of funding, and maximize the return on scientific investments. In order to support this, various data-sharing portals and repositories have been developed which not only provide searchable nanomaterial characterization data, but also provide access to standardized protocols for synthesis and characterization of nanomaterials as well as cutting-edge publications. The National Cancer Institute's (NCI) caNanoLab is a dedicated repository for all aspects pertaining to cancer-related nanotechnology data. The searchable database provides a unique opportunity for data mining and the use of artificial intelligence and machine learning, which aims to be an essential arm of future research studies, potentially speeding the design and optimization of next-generation therapies. It also provides an opportunity to track the latest trends and patterns in nanomedicine research. This manuscript provides the first look at such trends extracted from caNanoLab and compares these to similar metrics from the NCI's Nanotechnology Characterization Laboratory, a laboratory providing preclinical characterization of cancer nanotechnologies to researchers around the globe. Together, these analyses provide insight into the emerging interests of the research community and rise of promising nanoparticle technologies.


Subject(s)
Nanostructures , Neoplasms , United States , Humans , National Cancer Institute (U.S.) , Artificial Intelligence , Nanotechnology/methods , Nanomedicine/methods , Neoplasms/diagnosis , Neoplasms/drug therapy
7.
Pharmaceuticals (Basel) ; 15(7)2022 Jul 20.
Article in English | MEDLINE | ID: mdl-35890195

ABSTRACT

The earliest example of in vivo expression of exogenous mRNA is by direct intramuscular injection in mice without the aid of a delivery vehicle. The current state of the art for therapeutic nucleic acid delivery is lipid nanoparticles (LNP), which are composed of cholesterol, a helper lipid, a PEGylated lipid and an ionizable amine-containing lipid. The liver is the primary organ of LNP accumulation following intravenous administration and is also observed to varying degrees following intramuscular and subcutaneous routes. Delivery of nucleic acid to hepatocytes by LNP has therapeutic potential, but there are many disease indications that would benefit from non-hepatic LNP tissue and cell population targeting, such as cancer, and neurological, cardiovascular and infectious diseases. This review will concentrate on the current efforts to develop the next generation of tissue-targeted LNP constructs for therapeutic nucleic acids.

8.
Biomolecules ; 11(12)2021 11 25.
Article in English | MEDLINE | ID: mdl-34944412

ABSTRACT

Survival from pancreatic cancer remains extremely poor, in part because this malignancy is not diagnosed in the early stages, and precancerous pancreatic intraepithelial neoplasia (PanIN) lesions are not seen on routine radiographic imaging. Since the cholecystokinin-B receptor (CCK-BR) becomes over-expressed in PanIN lesions, it may serve as a target for early detection. We developed a biodegradable fluorescent polyplex nanoparticle (NP) that selectively targets the CCK-BR. The NP was complexed to a fluorescent oligonucleotide with Alexa Fluor 647 for far-red imaging and to an oligonucleotide conjugated to Alexa Fluor 488 for localization by immunohistochemistry. Fluorescence was detected over the pancreas of five- to ten-month-old LSL-KrasG12D/+; P48-Cre (KC) mice only after the injection of the receptor target-specific NP and not after injection of untargeted NP. Ex vivo tissue imaging and selective immunohistochemistry confirmed particle localization only to PanIN lesions in the pancreas and not in other organs, supporting the tissue specificity. A human pancreas tissue microarray demonstrated immunoreactivity for the CCK-BR only in the PanIN lesions and not in normal pancreas tissue. The long-term goal would be to develop this imaging tool for screening human subjects at high risk for pancreatic cancer to enable early cancer detection.


Subject(s)
Fluoresceins/administration & dosage , Optical Imaging/methods , Pancreatic Neoplasms/diagnostic imaging , Precancerous Conditions/diagnostic imaging , Receptor, Cholecystokinin B/metabolism , Sulfonic Acids/administration & dosage , Animals , Cell Line, Tumor , Disease Models, Animal , Early Detection of Cancer , Female , Humans , Immunohistochemistry , Male , Mice , Nanoparticles , Organ Specificity , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Protein Array Analysis , Proto-Oncogene Proteins p21(ras)/genetics
9.
Biomaterials ; 278: 121140, 2021 11.
Article in English | MEDLINE | ID: mdl-34634661

ABSTRACT

The in vivo fate of nanoformulated drugs is governed by the physicochemical properties of the drug and the functionality of nanocarriers. Nanoformulations such as polymeric micelles, which physically encapsulate poorly soluble drugs, release their payload into the bloodstream during systemic circulation. This results in three distinct fractions of the drug-nanomedicine: encapsulated, protein-bound, and free drug. Having a thorough understanding of the pharmacokinetic (PK) profiles of each fraction is essential to elucidate mechanisms of nanomedicine-driven changes in drug exposure and PK/PD relationships pharmacodynamic activity. Here, we present a comprehensive preclinical assessment of the poly (2-oxazoline)-based polymeric micelle of paclitaxel (PTX) (POXOL hl-PM), including bioequivalence comparison to the clinically approved paclitaxel nanomedicine, Abraxane®. Physicochemical characterization and toxicity analysis of POXOL hl-PM was conducted using standardized protocols by the Nanotechnology Characterization Laboratory (NCL). The bioequivalence of POXOL hl-PM to Abraxane® was evaluated in rats and rhesus macaques using the NCL's established stable isotope tracer ultrafiltration assay (SITUA) to delineate the plasma PK of each PTX fraction. The SITUA study revealed that POXOL hl-PM and Abraxane® had comparable PK profiles not only for total PTX but also for the distinct drug fractions, suggesting bioequivalence in given animal models. The comprehensive preclinical evaluation of POXOL hl-PM in this study showcases a series of widely applicable standardized studies by NCL for assessing nanoformulations prior to clinical investigation.


Subject(s)
Antineoplastic Agents, Phytogenic , Paclitaxel , Albumin-Bound Paclitaxel , Animals , Cell Line, Tumor , Drug Carriers , Isotopes , Macaca mulatta , Micelles , Rats , Rodentia , Therapeutic Equivalency
10.
Article in English | MEDLINE | ID: mdl-32830448

ABSTRACT

Despite imaging agents being some of the earliest nanomedicines in clinical use, the vast majority of current research and translational activities in the nanomedicine field involves therapeutics, while imaging agents are severely underrepresented. The reasons for this lack of representation are several fold, including difficulties in synthesis and scale-up, biocompatibility issues, lack of suitable tissue/disease selective targeting ligands and receptors, and a high bar for regulatory approval. The recent focus on immunotherapies and personalized medicine, and development of nanoparticle constructs with better tissue distribution and selectivity, provide new opportunities for nanomedicine imaging agent development. This manuscript will provide an overview of trends in imaging nanomedicine characterization and biocompatibility, and new horizons for future development. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.


Subject(s)
Diagnostic Imaging , Nanoparticles , Nanostructures , Biology , Nanomedicine , Precision Medicine
11.
Mol Pharm ; 17(10): 3794-3812, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32841040

ABSTRACT

We have developed a macromolecular prodrug platform based on poly(l-lysine succinylated) (PLS) that targets scavenger receptor A1 (SR-A1), a receptor expressed by myeloid and endothelial cells. We demonstrate the selective uptake of PLS by murine macrophage, RAW 264.7 cells, which was eliminated upon cotreatment with the SR-A inhibitor polyinosinic acid (poly I). Further, we observed no uptake of PLS in an SR-A1-deficient RAW 264.7 cell line, even after 24 h incubation. In mice, PLS distributed to lymphatic organs following i.v. injection, as observed by ex vivo fluorescent imaging, and accumulated in lymph nodes following both i.v. and i.d. administrations, based on immunohistochemical analysis with high-resolution microscopy. As a proof-of-concept, the HIV antiviral emtricitabine (FTC) was conjugated to the polymer's succinyl groups via ester bonds, with a drug loading of 14.2% (wt/wt). The prodrug (PLS-FTC) demonstrated controlled release properties in vitro with a release half-life of 15 h in human plasma and 29 h in esterase-inhibited plasma, indicating that drug release occurs through both enzymatic and nonenzymatic mechanisms. Upon incubation of PLS-FTC with human peripheral blood mononuclear cells (PBMCs), the released drug was converted to the active metabolite FTC triphosphate. In a pharmacokinetic study in rats, the prodrug achieved ∼7-19-fold higher concentrations in lymphatic tissues compared to those in FTC control, supporting lymphatic-targeted drug delivery. We believe that the SR-A1-targeted macromolecular PLS prodrug platform has extraordinary potential for the treatment of infectious diseases.


Subject(s)
Anti-HIV Agents/administration & dosage , Drug Carriers/chemistry , HIV Infections/drug therapy , Scavenger Receptors, Class A/metabolism , Animals , Anti-HIV Agents/pharmacokinetics , Drug Liberation , Emtricitabine/administration & dosage , Emtricitabine/pharmacokinetics , Female , Half-Life , Humans , Male , Mice , Poly I/pharmacology , Polylysine/chemistry , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Proof of Concept Study , RAW 264.7 Cells , Rats , Scavenger Receptors, Class A/antagonists & inhibitors , Scavenger Receptors, Class A/genetics
12.
Sci Adv ; 6(29): eaay9249, 2020 07.
Article in English | MEDLINE | ID: mdl-32832614

ABSTRACT

Nanoparticle (NP) delivery to solid tumors has recently been questioned. To better understand the magnitude of NP tumor delivery, we reanalyzed published murine NP tumor pharmacokinetic (PK) data used in the Wilhelm et al. study. Studies included in their analysis reporting matched tumor and blood concentration versus time data were evaluated using classical PK endpoints and compared to the unestablished percent injected dose (%ID) in tumor metric from the Wilhelm et al. study. The %ID in tumor was poorly correlated with standard PK metrics that describe NP tumor delivery (AUCtumor/AUCblood ratio) and only moderately associated with maximal tumor concentration. The relative tumor delivery of NPs was ~100-fold greater as assessed by the standard AUCtumor/AUCblood ratio than by %ID in tumor. These results strongly suggest that PK metrics and calculations can influence the interpretation of NP tumor delivery and stress the need to properly validate novel PK metrics against traditional approaches.

13.
ACS Pharmacol Transl Sci ; 3(3): 547-558, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32566919

ABSTRACT

The pharmacokinetics of nanomedicines are complicated by the unique dispositional characteristics of the drug carrier. Most simplistically, the carrier could be a solubilizing platform that allows administration of a hydrophobic drug. Alternatively, the carrier could be stable and release the drug in a controlled manner, allowing for distribution of the carrier to influence distribution of the encapsulated drug. A third potential dispositional mechanism is carriers that are not stably complexed to the drug, but rather bind the drug in a dynamic equilibrium, similar to the binding of unbound drug to protein; since the nanocarrier has distributional and binding characteristics unlike plasma proteins, the equilibrium binding of drug to a nanocarrier can affect pharmacokinetics in unexpected ways, diverging from classical protein binding paradigms. The recently developed stable isotope tracer ultrafiltration assay (SITUA) for nanomedicine fractionation is uniquely suited for distinguishing and comparing these carrier/drug interactions. Here we present the the encapsulated, unencapsulated, and unbound drug fraction pharmacokinetic profiles in rats for marketed nanomedicines, representing examples of controlled release (doxorubicin liposomes, Doxil; and doxorubicin HCl liposome generic), equilibrium binding (paclitaxel cremophor micelle solution, Taxol generic), and solubilizing (paclitaxel albumin nanoparticle, Abraxane; and paclitaxel polylactic acid micelle, Genexol-PM) nanomedicine formulations. The utility of the SITUA method in differentiating these unique pharmacokinetic profiles and its potential for use in establishing generic nanomedicine bioequivalence are discussed.

14.
Molecules ; 26(1)2020 Dec 31.
Article in English | MEDLINE | ID: mdl-33396545

ABSTRACT

The chloroquine family of antimalarials has a long history of use, spanning many decades. Despite this extensive clinical experience, novel applications, including use in autoimmune disorders, infectious disease, and cancer, have only recently been identified. While short term use of chloroquine or hydroxychloroquine is safe at traditional therapeutic doses in patients without predisposing conditions, administration of higher doses and for longer durations are associated with toxicity, including retinotoxicity. Additional liabilities of these medications include pharmacokinetic profiles that require extended dosing to achieve therapeutic tissue concentrations. To improve chloroquine therapy, researchers have turned toward nanomedicine reformulation of chloroquine and hydroxychloroquine to increase exposure of target tissues relative to off-target tissues, thereby improving the therapeutic index. This review highlights these reformulation efforts to date, identifying issues in experimental designs leading to ambiguity regarding the nanoformulation improvements and lack of thorough pharmacokinetics and safety evaluation. Gaps in our current understanding of these formulations, as well as recommendations for future formulation efforts, are presented.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Chloroquine/chemistry , Communicable Diseases/drug therapy , Drug Compounding/methods , Hydroxychloroquine/chemistry , Nanomedicine , Animals , Humans
16.
Mol Cancer Ther ; 18(3): 592-601, 2019 03.
Article in English | MEDLINE | ID: mdl-30587558

ABSTRACT

Photosensitizers can be integrated with drug delivery vehicles to develop chemophototherapy agents with antitumor synergy between chemo- and photocomponents. Long-circulating doxorubicin (Dox) in porphyrin-phospholipid (PoP) liposomes (LC-Dox-PoP) incorporates a phospholipid-like photosensitizer (2 mole %) in the bilayer of Dox-loaded stealth liposomes. Hematological effects of endotoxin-minimized LC-Dox-PoP were characterized via standardized assays. In vitro interaction with erythrocytes, platelets, and plasma coagulation cascade were generally unremarkable, whereas complement activation was found to be similar to that of commercial Doxil. Blood partitioning suggested that both the Dox and PoP components of LC-Dox-PoP were stably entrapped or incorporated in liposomes. This was further confirmed with pharmacokinetic studies in Fischer rats, which showed the PoP and Dox components of the liposomes both had nearly identical, long circulation half-lives (25-26 hours). In a large orthotopic mammary tumor model in Fischer rats, following intravenous dosing (2 mg/kg Dox), the depth of enhanced Dox delivery in response to 665 nm laser irradiation was over 1 cm. LC-Dox-PoP with laser treatment cured or potently suppressed tumor growth, with greater efficacy observed in tumors 0.8 to 1.2 cm, compared with larger ones. The skin at the treatment site healed within approximately 30 days. Taken together, these data provide insight into nanocharacterization and photo-ablation parameters for a chemophototherapy agent.


Subject(s)
Doxorubicin/analogs & derivatives , Drug Delivery Systems , Mammary Neoplasms, Animal/drug therapy , Phospholipids/pharmacology , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Disease Models, Animal , Doxorubicin/chemistry , Doxorubicin/pharmacology , Female , Humans , Mammary Neoplasms, Animal/chemistry , Mammary Neoplasms, Animal/pathology , Phospholipids/chemistry , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Rats , Rats, Inbred F344
17.
Article in English | MEDLINE | ID: mdl-29928669

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) remains the most aggressive malignancy with the lowest 5-year survival rate of all cancers in part owing to the lack of tumor-specific therapy and the rapid metastatic nature of this cancer. The gastrointestinal peptide gastrin is a trophic peptide that stimulates growth of PDAC in an autocrine fashion by interaction with the cholecystokinin receptor that is overexpressed in this malignancy. METHODS: We developed a therapeutic novel polyplex nanoparticle (NP) that selectively targets the cholecystokinin receptor on PDAC. The NP was characterized in vitro and stability testing was performed in human blood. The effects of the target-specific NP loaded with gastrin small interfering RNA (siRNA) was compared with an untargeted NP and with an NP loaded with a scrambled siRNA in vitro and in 2 orthotopic models of PDAC. A polymerase chain reaction metastasis array examined differentially expressed genes from control tumors compared with tumors of mice treated with the targeted polyplex NP. RESULTS: The polyplex NP forms a micelle that safely delivers specific gastrin siRNA to the tumor without off-target toxicity. Consistent with these findings, cellular uptake was confirmed only with the targeted fluorescently labeled NP by confocal microscopy in vitro and by IVIS fluorescent based imaging in mice bearing orthotopic pancreatic cancers but not found with untargeted NPs. Tumor uptake and release of the gastrin siRNA NP was verified by decreased cellular gastrin gene expression by quantitative reverse-transcription polymerase chain reaction and peptide expression by immunohistochemistry. Growth of PDAC was inhibited in a dose-related fashion in cell culture and in vivo. The targeted NP therapy completely blocked tumor metastasis and altered tumor-specific genes. CONCLUSIONS: Our polyplex nanoparticle platform establishes both a strong foundation for the development of receptor-targeted therapeutics and a unique approach for the delivery of siRNA in vivo, thus warranting further exploration of this approach in other types of cancers.

18.
Methods Mol Biol ; 1682: 135-147, 2018.
Article in English | MEDLINE | ID: mdl-29039099

ABSTRACT

NLRP3 inflammasome activation is one of the initial steps in an inflammatory cascade against pathogen/danger-associated molecular patterns (PAMPs/DAMPs), such as those arising from environmental toxins or nanoparticles, and is essential for innate immune response. NLRP3 inflammasome activation in cells can lead to the release of IL-1ß cytokine via caspase-1, which is required for inflammatory-induced programmed cell death (pyroptosis). Nanoparticles are commonly used as vaccine adjuvants and drug delivery vehicles to improve the efficacy and reduce the toxicity of chemotherapeutic agents. Several studies indicate that different nanoparticles (e.g., liposomes, polymer-based nanoparticles) can induce NLRP3 inflammasome activation. Generation of a pro-inflammatory response is beneficial for vaccine delivery to provide adaptive immunity, a necessary step for successful vaccination. However, similar immune responses for intravenously injected, drug-containing nanoparticles can result in immunotoxicity (e.g., silica nanoparticles). Evaluation of NLRP3-mediated inflammasome activation by nanoparticles may predict pro-inflammatory responses in order to determine if these effects may be mitigated for drug delivery or optimized for vaccine development. In this protocol, we outline steps to monitor the release of IL-1ß using PMA-primed THP-1 cells, a human monocytic leukemia cell line, as a model system. IL-1ß release is used as a marker of NLRP3 inflammasome activation.


Subject(s)
Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Nanoparticles/adverse effects , Cell Culture Techniques/methods , Cell Line , Cell Survival , Enzyme-Linked Immunosorbent Assay/methods , Humans , Interleukin-1beta/immunology
19.
Methods Mol Biol ; 1682: 211-219, 2018.
Article in English | MEDLINE | ID: mdl-29039105

ABSTRACT

Autophagy is a catabolic process involved in the degradation and recycling of long-lived proteins and damaged organelles for maintenance of cellular homeostasis, and it has also been proposed as a type II cell death pathway. The cytoplasmic components targeted for catabolism are enclosed in a double-membrane autophagosome that merges with lysosomes, to form autophagosomes, and are finally degraded by lysosomal enzymes. There is substantial evidence that several nanomaterials can cause autophagy and lysosomal dysfunction, either by prevention of autophagolysosome formation, biopersistence or inhibition of lysosomal enzymes. Such effects have emerged as a potential mechanism of cellular toxicity, which is also associated with various pathological conditions. In this chapter, we describe a method to monitor autophagy by fusion of the modifier protein MAP LC3 with green fluorescent protein (GFP; GFP-LC3). This method enables imaging of autophagosome formation in real time by fluorescence microscopy without perturbing the MAP LC3 protein function and the process of autophagy. With the GFP-LC3 protein fusion construct, a longitudinal study of autophagy can be performed in cells after treatment with nanomaterials.


Subject(s)
Autophagy , Green Fluorescent Proteins/analysis , Microscopy, Fluorescence/methods , Microtubule-Associated Proteins/analysis , Optical Imaging/methods , Animals , Green Fluorescent Proteins/genetics , LLC-PK1 Cells , Microtubule-Associated Proteins/genetics , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Swine , Transfection/methods
20.
Methods Mol Biol ; 1682: 223-239, 2018.
Article in English | MEDLINE | ID: mdl-29039106

ABSTRACT

An important step in the early development of a nanomedicine formulation is the evaluation of stability and drug release in biological matrices. Additionally, the measurement of encapsulated and unencapsulated nanomedicine drug fractions is important for the determination of bioequivalence (pharmacokinetic equivalence) of generic nanomedicines. Unfortunately, current methods to measure drug release in plasma are limited, and all have fundamental disadvantages including non-equilibrium conditions and process-induced artifacts. The primary limitation of current ultrafiltration (and equilibrium dialysis) methods for separation of encapsulated and unencapsulated drug and determination of drug release is the difficulty in accurately differentiating protein bound and encapsulated drug. Since the protein binding of most drugs is high (>70%) and can change in a concentration- and time-dependent manner, it is very difficult to accurately account for the fraction of non-filterable drug that is encapsulated within the nanomedicine and how much is bound to protein. The method in this chapter is an improvement of existing ultrafiltration protocols for nanomedicine fractionation in plasma, in which a stable isotope tracer is spiked into a nanomedicine containing plasma sample in order to precisely measure the degree of plasma protein binding. Determination of protein binding then allows for accurate calculation of encapsulated and unencapsulated nanomedicine drug fractions, as well as free and protein-bound fractions.


Subject(s)
Drug Liberation , Pharmaceutical Preparations/blood , Ultrafiltration/methods , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Antineoplastic Agents/metabolism , Blood Proteins/metabolism , Chromatography, High Pressure Liquid/methods , Docetaxel , Drug Carriers/chemistry , Humans , Isotopes/analysis , Liposomes/chemistry , Mass Spectrometry/methods , Nanostructures/chemistry , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/metabolism , Protein Binding , Taxoids/administration & dosage , Taxoids/blood , Taxoids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...