Proceedings Volume 10046

Visualizing and Quantifying Drug Distribution in Tissue

cover
Proceedings Volume 10046

Visualizing and Quantifying Drug Distribution in Tissue

Purchase the printed version of this volume at proceedings.com or access the digital version at SPIE Digital Library.

Volume Details

Date Published: 25 April 2017
Contents: 5 Sessions, 5 Papers, 10 Presentations
Conference: SPIE BiOS 2017
Volume Number: 10046

Table of Contents

icon_mobile_dropdown

Table of Contents

All links to SPIE Proceedings will open in the SPIE Digital Library. external link icon
View Session icon_mobile_dropdown
  • Front Matter: Volume 10046
  • Vibrational Spectroscopic Tracking of Drugs
  • Visualizing and Quantifying Drugs via Fluorescence
  • Advanced Imaging Tools for Quantifying Drug Uptake and Distribution
  • Multimodal, Light Scattering, Mass Spectrometry, and RNA-based Methods for Analyzing Drug Uptake and Effect
Front Matter: Volume 10046
icon_mobile_dropdown
Front Matter: Volume 10046
This PDF file contains the front matter associated with SPIE Proceedings Volume 10046, including the Title Page, Copyright information, Table of Contents, and Conference Committee listing.
Vibrational Spectroscopic Tracking of Drugs
icon_mobile_dropdown
Towards noninvasive drug distribution in tissues: coherent Raman microspectroscopy of chiral molecules
Georgi I. Petrov, Vladislav V. Yakovlev
Many biologically active molecules are chiral. Many drugs, which are currently in use, are supplied as an equimolar mixture of enantiomers. Although they have the same chemical structure, i.e. are not distinguishable by conventional Raman spectroscopy, most isomers of chiral drugs exhibit marked differences in biological activities such as pharmacology, toxicology, pharmacokinetics, metabolism, etc. In this report we introduced a new spectroscopic tool to extend nonlinear Raman spectroscopy to chiral substances.
Noninvasive label-free monitoring of cosmetics and pharmaceuticals in human skin using nonlinear optical microscopy (Conference Presentation)
Over the past decade, nonlinear optical microscopy has seen a dramatic rise in its use in research settings due to its noninvasiveness, enhanced penetration depth, intrinsic optical sectioning, and the ability to probe chemical compounds with molecular specificity without exogenous contrast agents. Nonlinear optical techniques including two-photon excitation fluorescence (2PEF), fluorescence lifetime imaging microscopy (FLIM), second harmonic generation (SHG), coherent anti-Stokes and stimulated Raman scattering (CARS and SRS, respectively), as well as transient and sum frequency absorption (TA and SFA, respectively), have been widely used to explore the physiology and microanatomy of skin. Recently, these modalities have shed light on dermal processes that could not have otherwise been observed, including the spatiotemporal monitoring of cosmetics and pharmaceuticals. However, a challenge quickly arises when studying such chemicals in a dermatological context: many exogenous compounds have optical signatures that can interfere with the signals that would otherwise be acquired from intact skin. For example, oily solvents exhibit strong signals when probing CH2 vibrations with CARS/SRS; chemical sun filters appear bright in 2PEF microscopy; and darkly colored compounds readily absorb light across a broad spectrum, producing strong TA/SFA signals. Thus, this discussion will first focus on the molecular contrast in skin that can be probed using the aforementioned nonlinear optical techniques. This will be followed by an overview of strategies that take advantage of the exogenous compounds’ optical signatures to probe spatiotemporal dynamics while preserving endogenous information from skin.
Surface-enhanced Raman scattering (SERS) imaging of alkyne-tagged small molecule drug in live cells with endocytosed gold nanoparticles
We propose the combination of alkyne-tag and surface-enhanced Raman scattering (SERS) spectroscopy to perform highly-sensitive and selective drug imaging in live cells. Gold nanoparticles are introduced in lysosomes through endocytosis as SERS agents, and the alkyne-tagged drugs are subsequently administered in cells. Raman microscopic observation reveals the arrival of drug in lysosome through enhanced Raman signal of alkyne. Since the peak of alkyne appears in Raman-silent region of biomolecules, selective detection of drugs is possible without background signal of endogenous molecules. From endocytosed gold nanoparticles in living HeLa cells, we observed distinct Raman signal from alkyne-tagged inhibitor of lysosomal enzyme.
Advanced imaging approaches for characterizing nanoparticle delivery and dispersion in skin (Conference Presentation)
Tarl W. Prow, Miko Yamada, Nhung Dang, et al.
The purpose of this research was to develop advanced imaging approaches to characterise the combination of elongated silica microparticles (EMP) and nanoparticles to control topical delivery of drugs and peptides. The microparticles penetrate through the epidermis and stop at the dermal-epidermal junction (DEJ). In this study we incorporated a fluorescent lipophilic dye, DiI, as a hydrophobic drug surrogate into the nanoparticle for visualization with microscopy. In another nanoparticle-based approach we utilized a chemically functionalized melanin nanoparticle for peptide delivery. These nanoparticles were imaged by coherent anti-Stoke Raman scattering (CARS) microscopy to characterize the delivery of these nanoparticles into freshly excised human skin. We compared four different coating approaches to combine EMP and nanoparticles. These data showed that a freeze-dried formulation with cross-linked alginate resulted in 100% of the detectable nanoparticle retained on the EMP. When this dry form of EMP-nanoparticle was applied to excised, living human abdominal skin, the EMP penetrated to the DEJ followed by controlled release of the nanoparticles. This formulation resulted in a sustained release profile, whereas a freeze-dried formulation without crosslinking showed an immediate burst-type release profile. These data show that advanced imaging techniques can give unique, label free data that shows promise for clinical investigations.
Visualizing and Quantifying Drugs via Fluorescence
icon_mobile_dropdown
Combining imaging with microbiopsy enables a more comprehensive approach for topical drug research (Conference Presentation)
Topical drug delivery is a challenging research field, but quantifying topical drug delivery also has significant challenges, especially in the clinical studies. Both cosmeceutical and pharmaceutical endpoints largely drive research in this area. Conventional drug delivery approaches primarily rely on testing trans dermal drug kinetics using excised skin in Franz cells. Thus is a largely unmet need for non- and minimally invasive approaches to evaluate topical drug delivery and efficacy in excised and volunteer skin. We are meeting this need through the development of non-invasive imaging based approaches such as fluorescent dermoscopy, fluorescence scanning and confocal microscopy followed by image analysis. Minimally invasive microbiopsies are being used to extract drug concentrations from tiny pieces of skin without the need for local anaesthetics and without scars. This combined strategy enables us to collect drug disposition information in addition to skin morphology and molecular characterisation which provides a more dynamic and comprehensive way to examine drug deliver, effects of enhancement technologies and efficacy.
Visualization of drug distribution of topical minocycline in human facial skin with fluorescence microscopy (Conference Presentation)
Maiko Hermsmeier, Tanvee Sawant, Diana Lac, et al.
Minocycline is an antibiotic regularly prescribed to treat acne vulgaris. The only commercially available minocycline comes in an oral dosage form, which often results in systemic adverse effects. A topical minocycline composition (BPX-01) was developed to provide localized and targeted delivery to the epidermis and pilosebaceous unit where acne-related bacteria, Propionibacterium acnes (P. acnes), reside. As minocycline is a known fluorophore, fluorescence microscopy was performed to investigate its potential use in visualizing minocycline distribution within tissues. BPX-01 with various concentrations of minocycline, was applied topically to freshly excised human facial skin specimens. Spatial distribution of minocycline and its fluorescence intensity within the stratum corneum, epidermis, dermis, and pilosebaceous unit were assessed. The resulting fluorescence intensity data as a function of minocycline concentration may indicate clinically relevant therapeutic doses of topical BPX-01 needed to kill P. acnes and reduce inflammation for successful clinical outcomes.
Quantitative imaging of intracellular signaling for personalized pancreatic cancer therapy in an in vivo avatar (Conference Presentation)
Kimberley S. Samkoe, Emily Schultz, Yeonjae Park, et al.
Pancreatic ductal adenocarcinomas (PDAC) are notoriously difficult to treat and in general, molecular targeted therapies have failed even when the targeted protein is overexpressed in the tumor tissue. Genetic mutations in extracellular receptors and downstream signaling proteins (i.e., RAS signaling pathway) and convoluted intracellular cross-talk between cell signaling pathways are likely reasons that these promising therapies fail. Monitoring the complex relationship between intracellular protein signaling is difficult and to-date, standard techniques that are used (Western blot, flow cytometry, immunohistochemistry, etc.) are invasive, static and do not accurately represent in vivo structure-function relationships. Here, we describe the development of an in ovo avatar using patient derived tumors grown on the chicken chorioallantoic membrane (CAM) and the novel fluorescence-based Quantitative Protein Expression Tracking (QUIET) methodology to bridge the gap between oncology, genomics and patient outcomes. Previously developed paired-agent imaging, was extended to a three-compartment model system in QUIET, which utilizes three types of imaging agents: novel fluorophore conjugated cell permeable targeted and untargeted small molecule paired-agents, in addition to a tumor perfusion agent that is not cell membrane permeable. We have demonstrated the ability to quantify the intracellular binding domain of a trans-membrane protein in vitro using cell permeable fluorescent agents (erlotinib-TRITC and control isotype-BODIPY FL). In addition, we have demonstrated imaging protocols to simultaneously image up to 6 spectrally distinct organic fluorophores in in ovo avatars using the Nuance EX (Perkin Elmer) and established proof-of-principle intracellular and extracellular protein concentrations of epidermal growth factor receptor using QUIET and traditional paired-agent imaging.
Advanced Imaging Tools for Quantifying Drug Uptake and Distribution
icon_mobile_dropdown
Direct visualization of functional heterogeneity in hepatobiliary metabolism
Chih-Ju Lin, Feng-Chieh Li, Yu-Yang Lee, et al.
Hepatycotes in the liver may appear similar in morphology, however, heterogeneities may exist in cellular metabolism. In this study, in vivo imaging of 6-carboxfluorescein diacetate (6-CFDA) metabolism in the liver was studied. We used two-photon fluorescence microscopy and hepatic window to provide quantification in studying hepatocellular metabolism. This model not only provides a potential platform for future study in hepatic responses and regulations, but also contributes to the fine-tuning of organ-specific functions so as to open up a new era of exciting discoveries.
Imaging bio-distribution of a topically applied dermatological cream on minipig skin using fluorescence lifetime imaging microscopy (Conference Presentation)
Aneesh Alex, Eric J. Chaney, Jennifer M. Criley, et al.
Currently there is a lack of in vivo techniques to evaluate the spatial bio-distribution of dermal drugs over time without the need to take multiple serial biopsies. To address this gap, we investigated the use of multi-photon optical imaging methods to non-invasively track drug distribution on miniature pig (Species: Sus scrofa, Strain: Göttingen) skin in vivo. Minipig skin is the standard comparative research model to human skin, and is anatomically and functionally similar. We employed fluorescence lifetime imaging microscopy (FLIM) to visualize the spatial distribution and residency time of a topically applied experimental dermatological cream. This was made possible by the endogenous fluorescent optical properties of the experimental drug (fluorescence lifetime > 3000 ps). Two different drug formulations were applied on 2 minipigs for 7 consecutive days, with the control creams applied on the contralateral side, followed by 7 days of post-application monitoring using a multi-modal optical imaging system (MPTflex-CARS, JenLab, Germany). FLIM images were obtained from the treated regions 24 hr post-application from day 1 to day 14 that allowed visualization of cellular and sub-cellular features associated with different dermal layers non-invasively to a depth of 200 µm. Five punch biopsies per animal were obtained from the corresponding treated regions between days 8 and 14 for bioanalytical analysis and comparison with results obtained using FLIM. In conclusion, utilization of non-invasive optical biopsy methods for dermal drug evaluation can provide true longitudinal monitoring of drug spatial distribution, remove sampling limitations, and be more time-efficient compared to traditional methods.
Multimodal, Light Scattering, Mass Spectrometry, and RNA-based Methods for Analyzing Drug Uptake and Effect
icon_mobile_dropdown
Identification and localization of trauma-related biomarkers using matrix assisted laser desorption/ionization imaging mass spectrometry
Current treatments for ocular and optic nerve trauma are largely ineffective and may have adverse side effects; therefore, new approaches are needed to understand trauma mechanisms. Identification of trauma-related biomarkers may yield insights into the molecular aspects of tissue trauma that can contribute to the development of better diagnostics and treatments. The conventional approach for protein biomarker measurement largely relies on immunoaffinity methods that typically can only be applied to analytes for which antibodies or other targeting means are available. Matrix assisted laser-assisted desorption/ionization imaging mass spectrometry (MALDI-IMS) is a specialized application of mass spectrometry that not only is well suited to the discovery of novel or unanticipated biomarkers, but also provides information about the spatial localization of biomarkers in tissue. We have been using MALDI-IMS to find traumarelated protein biomarkers in retina and optic nerve tissue from animal models subjected to ocular injury produced by either blast overpressure or mechanical torsion. Work to date by our group, using MALDI-IMS, found that the pattern of protein expression is modified in the injured ocular tissue as soon as 24 hr post-injury, compared to controls. Specific proteins may be up- or down-regulated by trauma, suggesting different tissue responses to a given injury. Ongoing work is directed at identifying the proteins affected and mapping their expression in the ocular tissue, anticipating that systematic analysis can be used to identify targets for prospective therapies for ocular trauma.
Miniaturizing 3D assay for high-throughput drug and genetic screens for small patient-derived tumor samples (Conference Presentation)
Asaf Rotem, Levi Garraway, Mei-Ju Su, et al.
Three-dimensional growth conditions reflect the natural environment of cancer cells and are crucial to be performed at drug screens. We developed a 3D assay for cellular transformation that involves growth in low attachment (GILA) conditions and is strongly correlated with the 50-year old benchmark assay-soft agar. Using GILA, we performed high-throughput screens for drugs and genes that selectively inhibit or increase transformation, but not proliferation. This phenotypic approach is complementary to our genetic approach that utilizes single-cell RNA-sequencing of a patient sample to identify putative oncogenes that confer sensitivity to drugs designed to specifically inhibit the identified oncoprotein. Currently, we are dealing with a big challenge in our field- the limited number of cells that might be extracted from a biopsy. Small patient-derived samples are hard to test in the traditional multiwell plate and it will be helpful to minimize the culture area and the experimental system. We managed to design a suitable microfluidic device for limited number of cells and perform the assay using image analysis. We aim to test drugs on tumor cells, outside of the patient body- and recommend on the ideal treatment that is tailored to the individual. This device will help to minimize biopsy-sampling volumes and minimize interventions in the patient’s tumor.
In vivo optical coherence tomography imaging of dissolution of hyaluronic acid microneedles in human skin (Conference Presentation)
Seungri Song, Jung Dong Kim, Jung-hyun Bae, et al.
Transdermal drug delivery (TDD) has been recently highlighted as an alternative to oral delivery and hypodermic injections. Among many methods, drug delivery using a microneedle (MN) is one of the promising administration strategies due to its high skin permeability, mininal invasiveness, and ease of injection. In addition, microneedle-based TDD is explored for cosmetic and therapeutic purposes, rapidly developing market of microneedle industry for general population. To date, visualization of microneedles inserted into biological tissue has primarily been performed ex vivo. MRI, CT and ultrasound imaging do not provide sufficient spatial resolution, and optical microscopy is not suitable because of their limited imaging depth; structure of microneedles located in 0.2~1mm into the skin cannot be visulalized. Optical coherence tomography (OCT) is a non-invasive, cross-sectional optical imaging modality for biological tissue with high spatial resolution and acquisition speed. Compared with ultrasound imaging, it exhibits superior spatial resolution (1~10 um) and high sensitivity, while providing an imaging depth of biological tissue down to 1~2 mm. Here, we present in situ imaging and analysis of the penetration and dissolution characteristics of hyaluronic acid based MNs (HA-MN) with various needle heights in human skin in vivo. In contrast to other studies, we measured the actual penetration depths of the HA-MNs by considering the experimentally measured refractive index of HA in the solid state. For the dissolution dynamics of the HA-MNs, time-lapse structural alteration of the MNs could be clearly visualized, and the volumetric changes of the MNs were measured with an image analysis algorithm.