Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biosens Bioelectron ; 236: 115408, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37267688

ABSTRACT

Microneedle sensors could enable minimally-invasive, continuous molecular monitoring - informing on disease status and treatment in real-time. Wearable sensors for pharmaceuticals, for example, would create opportunities for treatments personalized to individual pharmacokinetics. Here, we demonstrate a commercial-off-the-shelf (COTS) approach for microneedle sensing using an electrochemical aptamer-based sensor that detects the high-toxicity antibiotic, vancomycin. Wearable monitoring of vancomycin could improve patient care by allowing targeted drug dosing within its narrow clinical window of safety and efficacy. To produce sensors, we miniaturize the electrochemical aptamer-based sensors to a microelectrode format, and embed them within stainless steel microneedles (sourced from commercial insulin pen needles). The microneedle sensors achieve quantitative measurements in body-temperature undiluted blood. Further, the sensors effectively maintain electrochemical signal within porcine skin. This COTS approach requires no cleanroom fabrication or specialized equipment, and produces individually-addressable, sterilizable microneedle sensors capable of easily penetrating the skin. In the future, this approach could be adapted for multiplexed detection, enabling real-time monitoring of a range of biomarkers.


Subject(s)
Biosensing Techniques , Needles , Animals , Swine , Stainless Steel , Vancomycin , Skin , Oligonucleotides
2.
ACS Sens ; 8(1): 150-157, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36534756

ABSTRACT

Dose-limiting toxicity and significant patient-to-patient pharmacokinetic variability often render it difficult to achieve the safe and effective dosing of drugs. This is further compounded by the slow, cumbersome nature of the analytical methods used to monitor patient-specific pharmacokinetics, which inevitably rely on blood draws followed by post-facto laboratory analysis. Motivated by the pressing need for improved "therapeutic drug monitoring", we are developing electrochemical aptamer-based (EAB) sensors, a minimally invasive biosensor architecture that can provide real-time, seconds-resolved measurements of drug levels in situ in the living body. A key advantage of EAB sensors is that they are generalizable to the detection of a wide range of therapeutic agents because they are independent of the chemical or enzymatic reactivity of their targets. Three of the four therapeutic drug classes that have, to date, been shown measurable using in vivo EAB sensors, however, bind to nucleic acids as part of their mode of action, leaving open questions regarding the extent to which the approach can be generalized to therapeutics that do not. Here, we demonstrate real-time, in vivo measurements of plasma methotrexate, an antimetabolite (a mode of action not reliant on DNA binding) chemotherapeutic, following human-relevant dosing in a live rat animal model. By providing hundreds of drug concentration values, the resulting seconds-resolved measurements succeed in defining key pharmacokinetic parameters, including the drug's elimination rate, peak plasma concentration, and exposure (area under the curve), with unprecedented 5 to 10% precision. With this level of precision, we easily identify significant (>2-fold) differences in drug exposure occurring between even healthy rats given the same mass-adjusted methotrexate dose. By providing a real-time, seconds-resolved window into methotrexate pharmacokinetics, such measurements can be used to precisely "individualize" the dosing of this significantly toxic yet vitally important chemotherapeutic.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , Nucleic Acids , Humans , Rats , Animals , Methotrexate , Biosensing Techniques/methods , Drug Monitoring/methods
3.
ACS Sens ; 7(10): 2823-2832, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36205360

ABSTRACT

The continuous, real-time measurement of specific molecules in situ in the body would greatly improve our ability to understand, diagnose, and treat disease. The vast majority of continuous molecular sensing technologies, however, either (1) rely on the chemical or enzymatic reactivity of their targets, sharply limiting their scope, or (2) have never been shown (and likely will never be shown) to operate in the complex environments found in vivo. Against this background, here we review electrochemical aptamer-based (EAB) sensors, an electrochemical approach to real-time molecular monitoring that has now seen 15 years of academic development. The strengths of the EAB platform are significant: to date it is the only molecular measurement technology that (1) functions independently of the chemical reactivity of its targets, and is thus general, and (2) supports in vivo measurements. Specifically, using EAB sensors we, and others, have already reported the real-time, seconds-resolved measurements of multiple, unrelated drugs and metabolites in situ in the veins and tissues of live animals. Against these strengths, we detail the platform's remaining weaknesses, which include still limited measurement duration (hours, rather than the more desirable days) and the difficulty in obtaining sufficiently high performance aptamers against new targets, before then detailing promising approaches overcoming these hurdles. Finally, we close by exploring the opportunities we believe this potentially revolutionary technology (as well as a few, possibly competing, technologies) will create for both researchers and clinicians.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , Animals , Aptamers, Nucleotide/chemistry , Electrochemical Techniques
4.
Sci Rep ; 12(1): 5535, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35365672

ABSTRACT

Electrochemical aptamer-based (EAB) sensors support the real-time, high frequency measurement of pharmaceuticals and metabolites in-situ in the living body, rendering them a potentially powerful technology for both research and clinical applications. Here we explore quantification using EAB sensors, examining the impact of media selection and temperature on measurement performance. Using freshly-collected, undiluted whole blood at body temperature as both our calibration and measurement conditions, we demonstrate accuracy of better than ± 10% for the measurement of our test bed drug, vancomycin. Comparing titrations collected at room and body temperature, we find that matching the temperature of calibration curve collection to the temperature used during measurements improves quantification by reducing differences in sensor gain and binding curve midpoint. We likewise find that, because blood age impacts the sensor response, calibrating in freshly collected blood can improve quantification. Finally, we demonstrate the use of non-blood proxy media to achieve calibration without the need to collect fresh whole blood.


Subject(s)
Aptamers, Nucleotide , Calibration , Vancomycin
5.
ACS Sens ; 6(9): 3340-3347, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34491055

ABSTRACT

The ability to monitor drugs, metabolites, hormones, and other biomarkers in situ in the body would greatly advance both clinical practice and biomedical research. To this end, we are developing electrochemical aptamer-based (EAB) sensors, a platform technology able to perform real-time, in vivo monitoring of specific molecules irrespective of their chemical or enzymatic reactivity. An important obstacle to the deployment of EAB sensors in the challenging environments found in the living body is signal drift, whereby the sensor signal decreases over time. To date, we have demonstrated a number of approaches by which this drift can be corrected sufficiently well to achieve good measurement precision over multihour in vivo deployments. To achieve a much longer in vivo measurement duration, however, will likely require that we understand and address the sources of this effect. In response, here, we have systematically examined the mechanisms underlying the drift seen when EAB sensors and simpler, EAB-like devices are challenged in vitro at 37 °C in whole blood as a proxy for in vivo conditions. Our results demonstrate that electrochemically driven desorption of a self-assembled monolayer and fouling by blood components are the two primary sources of signal loss under these conditions, suggesting targeted approaches to remediating this degradation and thus improving the stability of EAB sensors and other, similar electrochemical biosensor technologies when deployed in the body.

6.
ACS Sens ; 6(6): 2299-2306, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34038076

ABSTRACT

Electrochemical aptamer-based sensors enable real-time molecular measurements in the living body. The spatial resolution of these measurements and ability to perform measurements in targeted locations, however, is limited by the length and width of the device's working electrode. Historically, achieving good signal to noise in the complex, noisy in vivo environment has required working electrode lengths of 3-6 mm. To enable sensor miniaturization, here we have enhanced the signaling current obtained for a sensor of given macroscopic dimensions by increasing its surface area. Specifically, we produced nanoporous gold via an electrochemical alloying/dealloying technique to increase the microscopic surface area of our working electrodes by up to 100-fold. Using this approach, we have miniaturized in vivo electrochemical aptamer-based (EAB) sensors (here using sensors against the antibiotic, vancomycin) by a factor of 6 while retaining sensor signal and response times. Conveniently, the fabrication of nanoporous gold is simple, parallelizable, and compatible with both two- and three-dimensional electrode architectures, suggesting that it may be of value to a range of electrochemical biosensor applications.


Subject(s)
Aptamers, Nucleotide , Nanopores , Electrochemical Techniques , Gold , Miniaturization
7.
PLoS One ; 16(3): e0249458, 2021.
Article in English | MEDLINE | ID: mdl-33765034

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0175933.].

8.
Anal Chem ; 92(20): 14063-14068, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32959647

ABSTRACT

Recent years have seen the development of a number of biosensor architectures that rely on target binding-induced changes in the rate of electron transfer from an electrode-bound receptor. Most often, the interrogation of these sensors has relied on voltammetric methods, such as square-wave voltammetry, which limit their time resolution to a few seconds. Here, we describe the use of an impedance-based approach, which we have termed electrochemical phase interrogation, as a means of collecting high time resolution measurements with sensors in this class. Specifically, using changes in the electrochemical phase to monitor target binding in an electrochemical-aptamer based (EAB) sensor, we achieve subsecond temporal resolution and multihour stability in measurements performed directly in undiluted whole blood. Electrochemical phase interrogation also offers improved insights into EAB sensors' signaling mechanism. By modeling the interfacial resistance and capacitance using equivalent circuits, we find that the only parameter that is altered by target binding is the charge-transfer resistance. This confirms previous claims that binding-induced changes in electron-transfer kinetics drive signaling in this class of sensors. Considering that a wide range of electrochemical biosensor architectures rely on this signaling mechanism, we believe that electrochemical phase interrogation may prove generalizable toward subsecond measurements of molecular targets.


Subject(s)
Aptamers, Nucleotide/chemistry , Tobramycin/blood , Biosensing Techniques/instrumentation , Biosensing Techniques/methods , Electrochemical Techniques , Hexanols/chemistry , Kinetics , Models, Chemical , Molecular Conformation , Oxidation-Reduction , Sulfhydryl Compounds/chemistry , Surface Properties
9.
Electrophoresis ; 40(5): 792-798, 2019 03.
Article in English | MEDLINE | ID: mdl-30597594

ABSTRACT

Spatial confinement, within cells or micro- and nanofabricated devices, impacts the conformation and binding kinetics of biomolecules. Understanding the role of spatial confinement on molecular behavior is important for comprehending diverse biological phenomena, as well as for designing biosensors. Specifically, the behavior of molecular binding under an applied electric field is of importance in the development of electrokinetic biosensors. Here, we investigate whether confinement of DNA oligomers in capillary electrophoresis impacts the binding kinetics of the DNA. To infer the role of confinement on hybridization dynamics, we perform capillary electrophoresis measurements on DNA oligomers within micro- and nanochannels, then apply first-order reaction dynamics theory to extract kinetic parameters from electropherogram data. We find that the apparent dissociation constants at the nanoscale (i.e., within a 100 nm channel) are lower than at the microscale (i.e., within a 1 µm channel), indicating stronger binding with increased confinement. This confirms, for the first time, that confinement-based enhancement of DNA hybridization persists under application of an electric field.


Subject(s)
DNA , Electrochemical Techniques/instrumentation , Microfluidic Analytical Techniques/instrumentation , Nanotechnology/instrumentation , Nucleic Acid Hybridization/methods , DNA/analysis , DNA/chemistry , DNA/metabolism , Equipment Design , Kinetics , Sodium Chloride/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...