Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Int J Pharm ; 658: 124209, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38718973

ABSTRACT

The USP Rotating Basket Dissolution Testing Apparatus 1 is listed in the USP as one of the tools to assess dissolution of oral solid dosage forms. Baskets of different mesh sizes can be used to differentiate between dissolution profiles of different formulations. Here, we used Particle Image Velocimetry (PIV) to study the hydrodynamics of the USP Apparatus 1 using baskets with different mesh openings (10-, 20- and 40-mesh) revolving at 100 rpm, when the vessel was filled with 500 mL. The velocity profiles throughout the liquid were found to vary significantly using baskets of different mesh sizes, typically increasing with increased size of the opening of the basket mesh, especially for axial and radial velocities. This, in turn, resulted in a significantly different flow rate through the basket, which can be expected to significantly impact the dissolution rate of the drug product. A comparison between the results of this work with those of a previous study with a 900-mL fill volume (Sirasitthichoke et al., Intern. J. Pharmaceutics, 2021, 607: 120976), shows that although the hydrodynamics in the USP Apparatus 1 changed with fill level in the vessel, the flow rate through the basket was not significantly affected. This implies that tablets dissolving in the two systems would experience similar tablet-liquid medium mass transfer coefficients, and therefore similar initial dissolution rates, but different dissolution profiles because of the difference in volume.


Subject(s)
Drug Liberation , Hydrodynamics , Rheology , Solubility , Tablets , Rheology/methods , Drug Compounding/methods , Drug Compounding/instrumentation , Chemistry, Pharmaceutical/methods , Particle Size , Technology, Pharmaceutical/methods
2.
AAPS J ; 25(6): 103, 2023 11 07.
Article in English | MEDLINE | ID: mdl-37936002

ABSTRACT

The in-person workshop "Drug Dissolution in Oral Drug Absorption" was held on May 23-24, 2023, in Baltimore, MD, USA. The workshop was organized into lectures and breakout sessions. Three common topics that were re-visited by various lecturers were amorphous solid dispersions (ASDs), dissolution/permeation interplay, and in vitro methods to predict in vivo biopharmaceutics performance and risk. Topics that repeatedly surfaced across breakout sessions were the following: (1) meaning and assessment of "dissolved drug," particularly of poorly water soluble drug in colloidal environments (e.g., fed conditions, ASDs); (2) potential limitations of a test that employs sink conditions for a poorly water soluble drug; (3) non-compendial methods (e.g., two-stage or multi-stage method, dissolution/permeation methods); (4) non-compendial conditions (e.g., apex vessels, non-sink conditions); and (5) potential benefit of having both a quality control method for batch release and a biopredictive/biorelevant method for biowaiver or bridging scenarios. An identified obstacle to non-compendial methods is the uncertainty of global regulatory acceptance of such methods.


Subject(s)
Biopharmaceutics , Intestinal Absorption , Humans , Drug Liberation , Solubility , Water
3.
AAPS J ; 25(1): 11, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36513860

ABSTRACT

This manuscript represents the view of the Dissolution Working Group of the IQ Consortium on the challenges of and recommendations on solubility measurements and development of dissolution methods for immediate release (IR) solid oral dosage forms formulated with amorphous solid dispersions. Nowadays, numerous compounds populate the industrial pipeline as promising drug candidates yet suffer from low aqueous solubility. In the oral drug product development process, solubility along with permeability is a key determinant to assure sufficient drug absorption along the intestinal tract. Formulating the drug candidate as an amorphous solid dispersion (ASD) is one potential option to address this issue. These formulations demonstrate the rapid onset of drug dissolution and can achieve supersaturated concentrations, which poses significant challenges to appropriately characterize solubility and develop quality control dissolution methods. This review strives to categorize the different dissolution and solubility challenges for ASD associated with 3 different topics: (i) definition of solubility and sink conditions for ASD dissolution, (ii) applications and development of non-sink dissolution (according to conventional definition) for ASD formulation screening and QC method development, and (iii) the advantages and disadvantages of using dissolution in detecting crystallinity in ASD formulations. Related to these challenges, successful examples of dissolution experiments in the context of control strategies are shared and may lead as an example for scientific consensus concerning dissolution testing of ASD.


Subject(s)
Solubility , Crystallization , Drug Liberation
4.
Int J Pharm ; 607: 120976, 2021 Sep 25.
Article in English | MEDLINE | ID: mdl-34363918

ABSTRACT

The USP Apparatus 1 (rotating basket), typically used to assess drug product reproducibility and evaluate oral solid dosage forms performance, consists of a cylindrical glass vessel with a hemispherical bottom and a wire basket rotating at constant speed. Baskets with different wire openings can be used in alternative to the standard mesh opening (40-mesh) in order to discriminate between drug formulations during early stage of drug product development. Any changes introduced by different basket geometries can potentially and significantly impact the system hydrodynamics and cause variability of results, thus affecting product quality. In this work, Particle Image Velocimetry (PIV) was used to experimentally quantify the velocity distribution in the USP rotating basket Apparatus 1 using baskets of different mesh sizes (10-, 20-, and 40-mesh size) under the typical operating conditions described in dissolution testing procedures. Similar flow patterns were observed in all cases. However, the radial and axial velocities in the USP Apparatus 1 generally increased with increasingly larger openings of the basket mesh. Increasing the basket agitation speed also resulted in an overall increase in the velocities, especially below in the innermost core region below the basket, where drug fragments typically reside. More importantly, the flow entering and leaving the baskets was quantified from the velocity profiles in the immediate vicinity of the baskets. It was found that the flow increased significantly with increasingly larger mesh openings, which can, in turn, promote faster dissolution of the oral solid dosage forms, thus affecting drug dissolution profiles. Hence, the selection of the basket mesh size must be carefully considered during drug product development.


Subject(s)
Hydrodynamics , Reproducibility of Results , Rheology , Solubility
5.
Mol Pharm ; 18(5): 2066-2081, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33784104

ABSTRACT

Dissolution of amorphous solid dispersions (ASD) can lead to the formation of amorphous drug-rich nano species (nanodroplets) via liquid-liquid phase separation or glass-liquid phase separation when the drug concentration exceeds the amorphous solubility. These nanodroplets have been shown to be beneficial for ASD performance both in vitro and in vivo. Thus, understanding the generation and stability of nanodroplets from ASD formulations is important. In this study, the impacts of polymer selection and active pharmaceutical ingredient (API) physicochemical properties (wet glass transition temperature (Tg) and log P) on nanodroplet release were studied. Six APIs with different physicochemical properties were formulated as ASDs with two polymers, polyvinylpyrrolidone/vinyl acetate (PVPVA) and hydroxypropyl methylcellulose acetate succinate (HPMCAS). Their release performance was evaluated using both powder and surface normalized dissolution of compacts. In general, HPMCAS-based dispersions resulted in higher drug release compared to PVPVA-based dispersions. The two polymers also exhibited different trends in nanodroplet formation as a function of drug loading (DL). PVPVA ASDs exhibited a "falling-off-the-cliff" effect, with a dramatic decline in release performance with a small increase in drug loading, while HPMCAS ASDs exhibited a negative "slope" in the release rate as a function of drug loading. For both polymers, low Tg compounds achieved higher levels of nanodroplet formation compared to high Tg compounds. The nanodroplets generated from ASD dissolution were also monitored with dynamic light scattering, and HPMCAS was found to be more effective at stabilizing nanodroplets against size increase. Insights from this study may be used to guide formulation design and selection of excipients based on API physicochemical properties.


Subject(s)
Excipients/chemistry , Pharmaceutical Preparations/chemistry , Chemistry, Pharmaceutical , Crystallization , Drug Compounding/methods , Drug Liberation , Methylcellulose/analogs & derivatives , Methylcellulose/chemistry , Nanoparticles/chemistry , Pyrrolidines/chemistry , Solubility , Transition Temperature , Vinyl Compounds/chemistry
6.
J Control Release ; 329: 316-327, 2021 01 10.
Article in English | MEDLINE | ID: mdl-33278481

ABSTRACT

Medical prescriptions for the alleviation of post-surgical pain are the most abundant source of opioids in circulation. As a systemic drug delivery source, opioids leave patients at high risk for side effects after being dosed. Given the significant rate of unauthorized use, distribution, addiction, and opioid related deaths, an alternative method of post-surgical analgesia is needed. Herein, we report the use of bio-resorbable poly(ester urea) (PEU) films that controllably deliver a non-opioid COX-2 inhibitor, etoricoxib, in vivo and in vitro as a model system for post-surgical pain control. PEU composition, drug-load, and film thickness were varied to selectively control etoricoxib elution. Elution data were fit to a Higuchi model, and the diffusion constant of etoricoxib was calculated in each of the films. Pharmacokinetic (pK) data from an in vivo rat model showed the local tissue concentration of etoricoxib at the study endpoint to be up to 23-fold higher in tissue then plasma. In a well-established mouse model of diabetic neuropathic pain in vivo film implantation showed effective relief of pain for more than 4 days post-implantation and efficacious local etoricoxib delivery. Overall, implementation of local drug delivery systems such as this could reduce the need for opioid prescriptions associated with current pain management strategies.


Subject(s)
Esters , Urea , Animals , Cyclooxygenase 2 Inhibitors/therapeutic use , Delayed-Action Preparations/therapeutic use , Double-Blind Method , Esters/therapeutic use , Etoricoxib/therapeutic use , Humans , Mice , Pain, Postoperative/drug therapy , Pyridines/therapeutic use , Rats , Sulfones/therapeutic use
7.
J Pharm Sci ; 109(11): 3471-3479, 2020 11.
Article in English | MEDLINE | ID: mdl-32888960

ABSTRACT

The physiological pH changes and peristalsis activities in gastrointestinal (GI) tract have big impact on the dissolution of oral drug products, when those oral drug products include APIs with pH-dependent solubility. It is well documented that predicting the bioperformance of those oral drug products can be challenging using compendial methods. To overcome this limitation, in vivo predictive dissolution apparatuses, such as the transfer model, have been developed to predict bioperformance of oral formulation candidates and drug products. In this manuscript we utilize a new transfer-model dissolution apparatus, the gastrointestinal simulator-α (GIS-α), to characterize its behavior in terms of transfer kinetics and pH, assess its reproducibility and adaptability to mimic different transfer conditions, as well as study dissolution of ketoconazole and dipyridamole as model BCS class IIb compounds. Availability of commercially available dissolution transfer systems with similar configuration to compendial dissolution apparatus, may be helpful to simplify and standardize in vivo predictive dissolution methodologies for BCS class IIb compounds in the future.


Subject(s)
Ketoconazole , Pharmaceutical Preparations , Administration, Oral , Dipyridamole , Hydrogen-Ion Concentration , Intestinal Absorption , Reproducibility of Results , Solubility
8.
J Pharm Sci ; 108(9): 3054-3062, 2019 09.
Article in English | MEDLINE | ID: mdl-31103787

ABSTRACT

In this article, experiments on tablets containing a model compound, grazoprevir, were conducted to explore how media selection for a quality control dissolution method can influence the sensitivity for the dissolution method toward drug crystallinity detection in an amorphous solid dispersion formulation. The experiment shows that under ideal nonsink conditions with respect to crystalline solubility, dissolution can indeed be predictive of crystallinity in the formulation. However, the limit of detection for crystallinity with quality control dissolution can change based on inherent variabilities in the drug product. In addition, it is demonstrated that the method's sensitivity and accuracy might be reduced if the crystalline particles are sufficiently small with respect to the solid dispersion particles. To further demonstrate the limits of the dissolution method, a dissolution model was also explored to simulate and predict the sensitivity of the dissolution response toward crystallinity based on solubility in the media and particle size of the crystals.


Subject(s)
Chemistry, Pharmaceutical/methods , Drug Compounding/standards , Models, Chemical , Quality Control , Quinoxalines/pharmacokinetics , Amides , Carbamates , Crystallization , Cyclopropanes , Drug Liberation , Feasibility Studies , Limit of Detection , Particle Size , Quinoxalines/chemistry , Solubility , Sulfonamides , Tablets
9.
AAPS J ; 21(3): 32, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30790200

ABSTRACT

This manuscript represents the perspective of the Dissolution Working Group of the International Consortium for Innovation and Quality in Pharmaceutical Development (IQ) and of two focus groups of the American Association of Pharmaceutical Scientists (AAPS): Process Analytical Technology (PAT) and In Vitro Release and Dissolution Testing (IVRDT). The intent of this manuscript is to show recent progress in the field of in vitro predictive dissolution modeling and to provide recommended general approaches to developing in vitro predictive dissolution models for both early- and late-stage formulation/process development and batch release. Different modeling approaches should be used at different stages of drug development based on product and process understanding available at those stages. Two industry case studies of current approaches used for modeling tablet dissolution are presented. These include examples of predictive model use for product development within the space explored during formulation and process optimization, as well as of dissolution models as surrogate tests in a regulatory filing. A review of an industry example of developing a dissolution model for real-time release testing (RTRt) and of academic case studies of enabling dissolution RTRt by near-infrared spectroscopy (NIRS) is also provided. These demonstrate multiple approaches for developing data-rich empirical models in the context of science- and risk-based process development to predict in vitro dissolution. Recommendations of modeling best practices are made, focused primarily on immediate-release (IR) oral delivery products for new drug applications. A general roadmap is presented for implementation of dissolution modeling for enhanced product understanding, robust control strategy, batch release testing, and flexibility toward post-approval changes.


Subject(s)
Chemistry, Pharmaceutical/methods , Drug Development/methods , Drug Liberation , Models, Biological , Administration, Oral , Capsules , Tablets
10.
AAPS J ; 21(2): 21, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30690680

ABSTRACT

This publication summarizes the proceedings and key outcomes of the first day ("Day 1") of the 3-day workshop on "Dissolution and Translational Modeling Strategies Enabling Patient-Centric Product Development." The overall aims of the workshop were to foster a productive dialog between industry and regulatory agencies and to discuss current strategies toward the development and implementation of clinically relevant dissolution specifications as an integral part of enhanced drug product understanding and effective drug product life-cycle management. The Day 1 podium presentations covered existing challenges and concerns for implementing highly valuable, yet often unique and novel experimental dissolution setups as quality control tools. In addition, several podium presentations highlighted opportunities to replace conventional dissolution testing with surrogate test methods to enable robust drug product and process understanding within the context of quality by design (QbD), new manufacturing technologies, and real-time release testing (RTRT). The topics covered on Day 1 laid the foundation for subsequent discussions which focused on the challenges related to establishing an in vitro-in vivo link and approaches for establishing clinically relevant drug product specifications which are becoming an expectation in regulatory submissions. Clarification of dissolution-related terminology used inconsistently among the scientific community, and the purpose of various testing approaches were key discussion topics of the Day 1 breakout sessions. The outcome of these discussions along with creative ways to overcome challenges related to bridging "exploratory dissolution approaches" with methods suitable for end-product control testing are captured within this report.


Subject(s)
Drug Development/methods , Quality Control , Animals , Congresses as Topic , Drug Development/standards , Humans , Solubility
11.
Int J Pharm ; 559: 147-155, 2019 Mar 25.
Article in English | MEDLINE | ID: mdl-30654058

ABSTRACT

Amorphous solid dispersions are a promising option for managing compounds with poor aqueous solubility. However, for compounds with high melting points, thermal stability limitations, or poor solubility in volatile solvents, conventional routes of hot melt extrusion or spray drying may not be viable. Co-precipitated amorphous dispersions (cPAD) can provide a solution. For the material studied in this paper, the cPAD material that was seemingly identical to spray dried material in terms of being single phase amorphous (as measured by DSC and XRD ) but showed slower dissolution behavior. It was identified that physical properties of the cPAD material could be improved to enhance wettability and improve dissolution performance. This was achieved by incorporating the cPAD material into a matrix of water soluble excipients generated via evaporative isolation routes. Importantly, this approach appears to offer another route to further increase the drug load in final dosage units and is significant as increased drug loading generally results in slower or incomplete release. Results showed successful proof of concept via in vitro biorelevant dissolution and confirmatory canine pharmacokinetic studies yielding comparable exposure for capsules comprised of conventional spray dried material as well as capsules with elevated drug load comprised of cPAD hierarchical particles.


Subject(s)
Pharmaceutical Preparations/chemistry , Animals , Chemistry, Pharmaceutical/methods , Desiccation/methods , Dogs , Drug Carriers/chemistry , Drug Compounding/methods , Drug Stability , Excipients/chemistry , Freezing , Polymers/chemistry , Solubility/drug effects , Wettability
12.
J Pharm Sci ; 108(1): 574-583, 2019 01.
Article in English | MEDLINE | ID: mdl-30395833

ABSTRACT

Solubility, dissolution, and precipitation in the gastrointestinal tract can be critical for the oral bioavailability of weakly basic drugs. To understand the dissolution and precipitation during the transfer out of the stomach into the intestine, a multicompartment transfer system was developed by modifying a conventional dissolution system. This transfer system included gastric, intestinal, sink and supersaturation, and reservoir compartments. Simulated gastric fluid and fasted state simulated intestinal fluid were used in the gastric and intestinal compartment, respectively, to mimic fasted condition. The new transfer system was evaluated based on 2 model weak bases, dipyridamole and ketoconazole. Traditional 2-stage dissolution using 250 mL of simulated gastric fluid media, followed by 250 mL of fasted state simulated intestinal fluid, was used as a reference methodology to compare dissolution and precipitation results. An in silico model was built using R software suite to simulate the in vitro time-dependent dissolution and precipitation process when formulations were tested using the transfer system. The precipitation rate estimated from the in vitro data was then used as the input for absorption and pharmacokinetic predictions using GastroPlus. The resultant simulated plasma concentration profiles were generally in good agreement with the observed clinical data, supporting the translatability of the transfer system in vitro precipitation kinetics to in vivo.


Subject(s)
Dipyridamole/pharmacokinetics , Gastrointestinal Tract/metabolism , Ketoconazole/pharmacokinetics , Administration, Oral , Chemical Precipitation , Computer Simulation , Drug Delivery Systems/methods , Humans , Hydrogen-Ion Concentration , Kinetics , Models, Biological , Solubility
13.
Int J Pharm ; 550(1-2): 418-428, 2018 Oct 25.
Article in English | MEDLINE | ID: mdl-30172750

ABSTRACT

Drug development is a long process which requires careful evaluation of the drug substance (active pharmaceutical ingredient, API), drug product (tablet, capsule etc.) and the bioperformance (both pre-clinical and clinical) before testing the efficacy of the final dosage form. The earliest assessment of a new drug substance requires an understanding of the safety and clinical performance (Phase 1) wherein faster processes (like on-site formulation strategy) have been set in place for quick clinical read-outs. One key gap that exists in this early assessment is the ability to evaluate modified release drug products. Here, an additive manufacturing approach is used to prepare polyvinyl alcohol (PVA) capsule shells using 3D printing (3DP), where the shells can be filled with either a solid or a liquid vehicle containing the API. In this work we demonstrate how we can delay the release of the API from the printed capsules allowing us to evaluate regional absorption in pre-clinical studies. By using 3DP, a new method to provide a series of release profiles is demonstrated, where the induction time of a delayed burst release is controlled by the wall thicknesses of printed capsules. New hanging baskets were also designed and 3D printed for the dissolution tests to better understand the rupturing of these capsules in an USP 2 dissolution apparatus. By controlling the wall thickness of the capsule, the induction time of drug release can be controlled from 12 to 198 min. This wide range of induction times demonstrated with this 3DP strategy is not currently available in a commercially available oral drug product form. Varying the induction times to the drug release to interrogate different regions of the GI tract is exhibited in vivo (beagle dogs) and initial analysis suggested a good in vitro/in vivo relationship (IVIVR).


Subject(s)
Capsules/administration & dosage , Intestinal Absorption , Printing, Three-Dimensional , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/chemistry , Capsules/chemistry , Carboxymethylcellulose Sodium/administration & dosage , Carboxymethylcellulose Sodium/chemistry , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Dogs , Drug Liberation , Gastrointestinal Tract/metabolism , Lamivudine/administration & dosage , Lamivudine/chemistry , Male , Polyvinyl Alcohol/administration & dosage , Polyvinyl Alcohol/chemistry
15.
J Pharm Sci ; 107(1): 34-41, 2018 01.
Article in English | MEDLINE | ID: mdl-29074376

ABSTRACT

This article intends to summarize the current views of the IQ Consortium Dissolution Working Group, which comprises various industry companies, on the roles of dissolution testing throughout pharmaceutical product development, registration, commercialization, and beyond. Over the past 3 decades, dissolution testing has evolved from a routine and straightforward test as a component of end-product release into a comprehensive set of tools that the developer can deploy at various stages of the product life cycle. The definitions of commonly used dissolution approaches, how they relate to one another and how they may be applied in modern drug development, and life cycle management is described in this article. Specifically, this article discusses the purpose, advantages, and limitations of quality control, biorelevant, and clinically relevant dissolution methods.


Subject(s)
Pharmaceutical Preparations/chemistry , Animals , Chemistry, Pharmaceutical/methods , Humans , Quality Control , Solubility
16.
AAPS J ; 19(6): 1537-1549, 2017 11.
Article in English | MEDLINE | ID: mdl-28831727

ABSTRACT

This manuscript represents the perspective of the Dissolution Analytical Working Group of the IQ Consortium. The intent of this manuscript is to highlight the challenges of, and to provide a recommendation on, the development of clinically relevant dissolution specifications (CRS) for immediate release (IR) solid oral dosage forms. A roadmap toward the development of CRS for IR products containing active ingredients with a non-narrow therapeutic window is discussed, within the context of mechanistic dissolution understanding, supported by in-human pharmacokinetic (PK) data. Two case studies present potential outcomes of following the CRS roadmap and setting dissolution specifications. These cases reveal some benefits and challenges of pursuing CRS with additional PK data, in light of current regulatory positions, including that of the US Food and Drug Administration (FDA), who generally favor this approach, but with the understanding that both industry and regulatory agency perspectives are still evolving in this relatively new field. The CRS roadmap discussed in this manuscript also describes a way to develop clinically relevant dissolution specifications based primarily on dissolution data for batches used in pivotal clinical studies, acknowledging that not all IR product development efforts need to be supported by additional PK studies, albeit with the associated risk of potentially unnecessarily tight manufacturing controls. Recommendations are provided on what stages during the life cycle investment into in vivo studies may be valuable. Finally, the opportunities for CRS within the context of post-approval changes, Modeling and Simulation (M&S), and the application of biowaivers, are briefly discussed.


Subject(s)
Solubility , Administration, Oral , Humans , Models, Biological , Tablets
17.
J Pharm Sci ; 104(9): 2913-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25611455

ABSTRACT

Although in vitro-in vivo correlations (IVIVCs) are commonly pursued for modified-release products, there are limited reports of successful IVIVCs for immediate-release (IR) formulations. This manuscript details the development of a Multiple Level C IVIVC for the amorphous solid dispersion formulation of suvorexant, a BCS class II compound, and its application to establishing dissolution specifications and in-process controls. Four different 40 mg batches were manufactured at different tablet hardnesses to produce distinct dissolution profiles. These batches were evaluated in a relative bioavailability clinical study in healthy volunteers. Although no differences were observed for the total exposure (AUC) of the different batches, a clear relationship between dissolution and Cmax was observed. A validated Multiple Level C IVIVC against Cmax was developed for the 10, 15, 20, 30, and 45 min dissolution time points and the tablet disintegration time. The relationship established between tablet tensile strength and dissolution was subsequently used to inform suitable tablet hardness ranges within acceptable Cmax limits. This is the first published report for a validated Multiple Level C IVIVC for an IR solid dispersion formulation demonstrating how this approach can facilitate Quality by Design in formulation development and help toward clinically relevant specifications and in-process controls.


Subject(s)
Azepines/chemistry , Azepines/pharmacokinetics , Tablets/chemistry , Tablets/pharmacokinetics , Triazoles/chemistry , Triazoles/pharmacokinetics , Area Under Curve , Biological Availability , Chemistry, Pharmaceutical/methods , Cross-Over Studies , Hardness , Healthy Volunteers , Humans , Solubility
18.
Front Behav Neurosci ; 6: 36, 2012.
Article in English | MEDLINE | ID: mdl-22783176

ABSTRACT

Dopamine projections that extend from the ventral tegmental area to the striatum have been implicated in the biological basis for behaviors associated with reward and addiction. Until recently, it has been difficult to evaluate the complex balance of energy utilization and neural activity in the striatum. Many techniques such as electrophysiology, functional magnetic resonance imaging (fMRI), and fast-scan cyclic voltammetry have been employed to monitor these neurochemical and neurophysiological changes. In this brain region, physiological responses to cues and rewards cause local, transient pH changes. Oxygen and pH are coupled in the brain through a complex system of blood flow and metabolism as a result of transient neural activity. Indeed, this balance is at the heart of imaging studies such as fMRI. To this end, we measured pH and O(2) changes with fast-scan cyclic voltammetry in the striatum as indices of changes in metabolism and blood flow in vivo in three Macaca mulatta monkeys during reward-based behaviors. Specifically, the animals were presented with Pavlovian conditioned cues that predicted different probabilities of liquid reward. They also received free reward without predictive cues. The primary detected change consisted of pH shifts in the striatal extracellular environment following the reward predicting cues or the free reward. We observed three types of cue responses that consisted of purely basic pH shifts, basic pH shifts followed by acidic pH shifts, and purely acidic pH shifts. These responses increased with reward probability, but were not significantly different from each other. The pH changes were accompanied by increases in extracellular O(2). The changes in pH and extracellular O(2) are consistent with current theories of metabolism and blood flow. However, they were of sufficient magnitude that they masked dopamine changes in the majority of cases. The findings suggest a role of these chemical responses in neuronal reward processing.

20.
Anal Chem ; 80(11): 4040-8, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18433146

ABSTRACT

Fast-scan cyclic voltammetry has been used in a variety of applications and has been shown to be especially useful to monitor chemical fluctuations of neurotransmitters such as dopamine within the mammalian brain. A major limitation of this procedure, however, is the large amplitude of the background current relative to the currents for the solution species of interest. Furthermore, the background tends to drift, and this drift limits the use of digital background subtraction techniques to intervals less than 90 s before distortion of dopamine signals occurs. To minimize the impact of the background, a procedure termed analog background subtraction is reported here. The background is recorded, and its inverse is played back to the current transducer during data acquisition so that it cancels the background in subsequent scans. Background drift still occurs and is recorded, but its magnitude is small compared to the original background. This approach has two advantages. First it allows the use of higher gains in the current transducer, minimizing quantization noise. Second, because the background amplitude is greatly reduced, principal component regression could be used to separate the contributions from drift, dopamine, and pH when appropriate calibrations were performed. We demonstrate the use of this approach with several applications. First, transient dopamine fluctuations were monitored for 15 min in a flowing injection apparatus. Second, evoked release of dopamine was monitored for a similar period in the brain of an anesthetized rat. Third, dopamine was monitored in the brain of freely moving rats over a 30 min interval. By analyzing the fluctuations in each resolved component, we were able to show that cocaine causes significant fluctuations in dopamine concentration in the brain while those for the background and pH remain unchanged from their predrug value.


Subject(s)
Dopamine/analysis , Analog-Digital Conversion , Anesthesia , Animals , Brain/metabolism , Brain/physiology , Carbon/chemistry , Carbon Fiber , Electrochemistry , Male , Microelectrodes , Principal Component Analysis , Rats , Rats, Sprague-Dawley , Time Factors , Transducers , Walking
SELECTION OF CITATIONS
SEARCH DETAIL
...