Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Garry Milman is active.

Publication


Featured researches published by Garry Milman.


Journal of Chromatography A | 2010

Simultaneous quantification of cannabinoids and metabolites in oral fluid by two-dimensional gas chromatography mass spectrometry.

Garry Milman; Allan J. Barnes; Ross H. Lowe; Marilyn A. Huestis

Development and validation of a method for simultaneous identification and quantification of Delta9-tetrahydrocannabinol (THC), cannabidiol (CBD), cannabinol (CBN), and metabolites 11-hydroxy-THC (11-OH-THC) and 11-nor-9-carboxy-THC (THCCOOH) in oral fluid. Simultaneous analysis was problematic due to different physicochemical characteristics and concentration ranges. Neutral analytes, such as THC and CBD, are present in ng/mL, rather than pg/mL concentrations, as observed for the acidic THCCOOH biomarker in oral fluid. THCCOOH is not present in cannabis smoke, definitively differentiating cannabis use from passive smoke exposure. THC, 11-OH-THC, THCCOOH, CBD, and CBN quantification was achieved in a single oral fluid specimen collected with the Quantisal device. One mL oral fluid/buffer solution (0.25 mL oral fluid and 0.75 mL buffer) was applied to conditioned CEREX Polycrom THC solid-phase extraction (SPE) columns. After washing, THC, 11-OH-THC, CBD, and CBN were eluted with hexane/acetone/ethyl acetate (60:30:20, v/v/v), derivatized with N,O-bis-(trimethylsilyl)trifluoroacetamide and quantified by two-dimensional gas chromatography electron ionization mass spectrometry (2D-GCMS) with cold trapping. Acidic THCCOOH was separately eluted with hexane/ethyl acetate/acetic acid (75:25:2.5, v/v/v), derivatized with trifluoroacetic anhydride and hexafluoroisopropanol, and quantified by the more sensitive 2D-GCMS-electron capture negative chemical ionization (NCI-MS). Linearity was 0.5-50 ng/mL for THC, 11-OH-THC, CBD and 1-50 ng/mL for CBN. The linear dynamic range for THCCOOH was 7.5-500 pg/mL. Intra- and inter-assay imprecision as percent RSD at three concentrations across the linear dynamic range were 0.3-6.6%. Analytical recovery was within 13.8% of target. This new SPE 2D-GCMS assay achieved efficient quantification of five cannabinoids in oral fluid, including pg/mL concentrations of THCCOOH by combining differential elution, 2D-GCMS with electron ionization and negative chemical ionization. This method will be applied to quantification of cannabinoids in oral fluid specimens from individuals participating in controlled cannabis and Sativex (50% THC and 50% CBD) administration studies, and during cannabis withdrawal.


Clinical Chemistry | 2011

Oral Fluid Cannabinoids in Chronic, Daily Cannabis Smokers during Sustained, Monitored Abstinence

Dayong Lee; Garry Milman; Allan J. Barnes; Robert S. Goodwin; Jussi Hirvonen; Marilyn A. Huestis

BACKGROUND Oral fluid (OF) is an accepted alternative biological matrix for drug treatment, workplace, and DUID (driving under the influence of drugs) investigations, but establishing the cannabinoid OF detection window and concentration cutoff criteria are important. METHODS Cannabinoid concentrations were quantified in OF from chronic, daily cannabis smokers during monitored abstinence. Δ(9)-tetrahydrocannabinol (THC)(3), cannabidiol (CBD), cannabinol (CBN), and 11-nor-9-carboxy-THC (THCCOOH) were determined in daily OF samples collected with the Quantisal™ device. GC-MS limits of quantification (LOQ) were 0.5 μg/L for THC and CBD, 1 μg/L for CBN, and 7.5 ng/L for THCCOOH. RESULTS After providing written informed consent for this institutional review board-approved study, 28 participants resided from 4 to 33 days on the secure research unit and provided 577 OF specimens. At the LOQ, THC was generally quantifiable for 48 h, whereas CBD and CBN were detected only at admission. Median THCCOOH detection time was 13 days (CI 6.4-19.6 days). Mean THC detection rates decreased from 89.3% at admission to 17.9% after 48 h, whereas THCCOOH gradually decreased from 89.3% to 64.3% within 4 days. Criteria of THC ≥2 μg/L and THCCOOH ≥20 ng/L reduced detection to <48 h in chronic cannabis smokers. An OF THCCOOH/THC ratio ≤4 ng/μg or presence of CBD or CBN may indicate more recent smoking. CONCLUSIONS THC, THCCOOH, CBD, and CBN quantification in confirmatory OF cannabinoid testing is recommended. Inclusion of multiple cannabinoid cutoffs accounted for residual cannabinoid excretion in OF from chronic, daily cannabis smokers and could reduce the potential for positive test results from passive cannabis smoke exposure and lead to greatly improved test interpretation.


Clinical Chemistry | 2012

Cannabinoid Disposition in Oral Fluid after Controlled Smoked Cannabis

Dayong Lee; David M. Schwope; Garry Milman; Allan J. Barnes; David A. Gorelick; Marilyn A. Huestis

BACKGROUND We measured Δ(9)-tetrahydrocannabinol (THC), 11-nor-9-carboxy-THC (THCCOOH), cannabidiol (CBD), and cannabinol (CBN) disposition in oral fluid (OF) following controlled cannabis smoking to evaluate whether monitoring multiple cannabinoids in OF improved OF test interpretation. METHODS Cannabis smokers provided written informed consent for this institutional review board-approved study. OF was collected with the Quantisal™ device following ad libitum smoking of one 6.8% THC cigarette. Cannabinoids were quantified by 2-dimensional GC-MS. We evaluated 8 alternative cutoffs based on different drug testing program needs. RESULTS 10 participants provided 86 OF samples -0.5 h before and 0.25, 0.5, 1, 2, 3, 4, 6, and 22 h after initiation of smoking. Before smoking, OF samples of 4 and 9 participants were positive for THC and THCCOOH, respectively, but none were positive for CBD and CBN. Maximum THC, CBD, and CBN concentrations occurred within 0.5 h, with medians of 644, 30.4, and 49.0 μg/L, respectively. All samples were THC positive at 6 h (2.1-44.4 μg/L), and 4 of 6 were positive at 22 h. CBD and CBN were positive only up to 6 h in 3 (0.6-2.1 μg/L) and 4 (1.0-4.4 μg/L) participants, respectively. The median maximum THCCOOH OF concentration was 115 ng/L, with all samples positive to 6 h (14.8-263 ng/L) and 5 of 6 positive at 22 h. CONCLUSIONS By quantifying multiple cannabinoids and evaluating different analytical cutoffs after controlled cannabis smoking, we determined windows of drug detection, found suggested markers of recent smoking, and minimized the potential for passive contamination.


Clinical Chemistry | 2012

Cannabinoid Stability in Authentic Oral Fluid after Controlled Cannabis Smoking

Dayong Lee; Garry Milman; David M. Schwope; Allan J. Barnes; David A. Gorelick; Marilyn A. Huestis

BACKGROUND Defining cannabinoid stability in authentic oral fluid (OF) is critically important for result interpretation. There are few published OF stability data, and of those available, all employed fortified synthetic OF solutions or elution buffers; none included authentic OF following controlled cannabis smoking. METHODS An expectorated OF pool and a pool of OF collected with Quantisal™ devices were prepared for each of 10 participants. Δ⁹-tetrahydrocannabinol (THC), 11-nor-9-carboxy-THC (THCCOOH), cannabidiol (CBD), and cannabinol (CBN) stability in each of 10 authentic expectorated and Quantisal-collected OF pools were determined after storage at 4 °C for 1 and 4 weeks and at -20 °C for 4 and 24 weeks. Results within ±20% of baseline concentrations analyzed within 24 h of collection were considered stable. RESULTS All Quantisal OF cannabinoid concentrations were stable for 1 week at 4 °C. After 4 weeks at 4 °C, as well as 4 and 24 weeks at -20 °C, THC was stable in 90%, 80%, and 80% and THCCOOH in 89%, 40%, and 50% of Quantisal samples, respectively. Cannabinoids in expectorated OF were less stable than in Quantisal samples when refrigerated or frozen. After 4 weeks at 4 and -20 °C, CBD and CBN were stable in 33%-100% of Quantisal and expectorated samples; by 24 weeks at -20 °C, CBD and CBN were stable in ≤ 44%. CONCLUSIONS Cannabinoid OF stability varied by analyte, collection method, and storage duration and temperature, and across participants. OF collection with a device containing an elution/stabilization buffer, sample storage at 4 °C, and analysis within 4 weeks is preferred to maximize result accuracy.


Clinica Chimica Acta | 2012

Cannabinoids and Metabolites in Expectorated Oral Fluid Following Controlled Smoked Cannabis

Garry Milman; David M. Schwope; David A. Gorelick; Marilyn A. Huestis

BACKGROUND ∆(9)-Tetrahydrocannabinol (THC) in oral fluid (OF) implies cannabis intake, but eliminating passive exposure and improving interpretation of test results requires additional research. METHODS Ten adult cannabis users smoked ad libitum one 6.8% THC cigarette. Expectorated OF was collected for up to 22 h, and analyzed within 24h of collection. THC, 11-nor-9-carboxy-THC (THCCOOH), cannabidiol, and cannabinol were quantified by 2-dimensional-GCMS. RESULTS Eighty specimens were analyzed; 6 could not be collected due to dry mouth. THC was quantifiable in 95.2%, cannabidiol in 69.3%, cannabinol in 62.3%, and THCCOOH in 94.7% of specimens. Highest THC, cannabidiol, and cannabinol concentrations were 22370, 1000, and 1964 μg/l, respectively, 0.25 h after the start of smoking; THCCOOH peaked within 2h (up to 560 ng/l). Concentrations 6h after smoking were THC (0.9-90.4 μg/l) and THCCOOH (17.0-151 ng/l) (8 of 9 positive for both); only 4 were positive for cannabidiol (0.5-2.4 μg/l) and cannabinol (1.0-3.0 μg/l). By 22 h, there were 4 THC (0.4-10.3 μg/l), 5 THCCOOH (6.0-24.0 ng/l), 1 cannabidiol (0.3 μg/l), and no cannabinol positive specimens. CONCLUSIONS THCCOOH in OF suggests no passive contamination, and CBD and CBN suggest recent cannabis smoking. Seventeen alternative cutoffs were evaluated to meet the needs of different drug testing programs.


Clinical Chemistry | 2010

Disposition of Cannabinoids in Oral Fluid after Controlled Around-the-Clock Oral THC Administration

Garry Milman; Allan J. Barnes; David M. Schwope; Eugene W. Schwilke; William D. Darwin; Robert S. Goodwin; Deanna L. Kelly; David A. Gorelick; Marilyn A. Huestis

BACKGROUND Oral fluid, a promising alternative matrix for drug monitoring in clinical and forensic investigations, offers noninvasive sample collection under direct observation. Cannabinoid distribution into oral fluid is complex and incompletely characterized due to the lack of controlled drug administration studies. METHODS To characterize cannabinoid disposition in oral fluid, we administered around-the-clock oral Delta(9)-tetrahydrocannabinol (THC) (Marinol) doses to 10 participants with current daily cannabis use. We obtained oral fluid samples (n=440) by use of Quantisal collection devices before, during, and after 37 20-mg THC doses over 9 days. Samples were extracted with multiple elution solvents from a single SPE column and analyzed by 2-dimensional GC-MS with electron-impact ionization for THC, 11-hydroxy-THC (11-OH-THC), cannabidiol, and cannabinol and negative chemical ionization for 11-nor-9-carboxy-THC (THCCOOH). Linear ranges were 0.5-50 microg/L, with the exception of cannabinol (1-50 microg/L) and THCCOOH (7.5-500 ng/L). RESULTS THCCOOH was the most prevalent analyte in 432 samples (98.2%), with concentrations up to 1117.9 ng/L. In contrast, 11-OH-THC was not identified in any sample; cannabidiol and cannabinol were quantified in 3 and 8 samples, respectively, with maximum concentrations of 2.1 and 13 microg/L. THC was present in only 20.7% of samples, with highest concentrations near admission (median 4.2 microg/L, range 0.6-481.9) from previously self-administered smoked cannabis. CONCLUSIONS Measurement of THCCOOH in OF not only identifies cannabis exposure, but also minimizes the possibility of passive inhalation. THCCOOH may be a better analyte for detection of cannabis use.


Clinical Chemistry | 2011

Oral Fluid and Plasma Cannabinoid Ratios after Around-the-Clock Controlled Oral Δ9-Tetrahydrocannabinol Administration

Garry Milman; David M. Schwope; Eugene W. Schwilke; William D. Darwin; Deanna L. Kelly; Robert S. Goodwin; David A. Gorelick; Marilyn A. Huestis

BACKGROUND Oral fluid (OF) testing is increasingly important for drug treatment, workplace, and drugged-driving programs. There is interest in predicting plasma or whole-blood concentrations from OF concentrations; however, the relationship between these matrices is incompletely characterized because of few controlled drug-administration studies. METHODS Ten male daily cannabis smokers received around-the-clock escalating 20-mg oral Δ(9)-tetrahydrocannabinol (THC, dronabinol) doses (40-120 mg/day) for 8 days. Plasma and OF samples were simultaneously collected before, during, and after dosing. OF THC, 11-hydroxy-THC and 11-nor-9-carboxy-THC (THCCOOH) were quantified by GC-MS at 0.5-μg/L, 0.5-μg/L, and 7.5-ng/L limits of quantification (LOQs), respectively. In plasma, the LOQs were 0.25 μg/L for THC and THCCOOH, and 0.5 μg/L for 11-hydroxy-THC. RESULTS Despite multiple oral THC administrations each day and increasing plasma THC concentrations, OF THC concentrations generally decreased over time, reflecting primarily previously self-administered smoked cannabis. The logarithms of the THC concentrations in oral fluid and plasma were not significantly correlated (r = -0.10; P = 0.065). The OF and plasma THCCOOH concentrations, albeit with 1000-fold higher concentrations in plasma, increased throughout dosing. The logarithms of OF and plasma THCCOOH concentrations were significantly correlated (r = 0.63; P < 0.001), although there was high interindividual variation. A high OF/plasma THC ratio and a high OF THC/THCCOOH ratio indicated recent cannabis smoking. CONCLUSIONS OF monitoring does not reliably detect oral dronabinol intake. The time courses of THC and THCCOOH concentrations in plasma and OF were different after repeated oral THC doses, and high interindividual variation was observed. For these reasons, OF cannabinoid concentrations cannot predict concurrent plasma concentrations.


Clinical Chemistry | 2010

Validation of an Enzyme Immunoassay for Detection and Semiquantification of Cannabinoids in Oral Fluid

David M. Schwope; Garry Milman; Marilyn A. Huestis

BACKGROUND Oral fluid (OF) is gaining prominence as an alternative matrix for monitoring drugs of abuse in the workplace, criminal justice, and driving under the influence of drugs programs. It is important to characterize assay performance and limitations of screening techniques for Delta(9)-tetrahydrocannabinol (THC) in OF. METHODS We collected OF specimens by use of the Quantisal OF collection device from 13 daily cannabis users after controlled oral cannabinoid administration. All specimens were tested with the Immunalysis Sweat/OF THC Direct ELISA and confirmed by 2-dimensional GC-MS. RESULTS The limit of detection was <1 microg/L THC equivalent, and the assay demonstrated linearity from 1 to 50 microg/L, with semiquantification to 200 microg/L. Intraplate imprecision (n = 7) ranged from 2.9% to 7.7% CV, and interplate imprecision (n = 20) was 3.0%-9.1%. Cross-reactivities at 4 microg/L were as follows: 11-hydroxy-THC, 198%; Delta(8)-tetrahydrocannabinol (Delta(8)-THC), 128%; 11-nor-9-carboxy-THC (THCCOOH), 121%; THC (target), 98%; cannabinol, 87%; THCCOOH-glucuronide, 11%; THC-glucuronide, 10%; and cannabidiol, 2.4%. Of 499 tested OF specimens, 52 confirmed positive (THC 2.0-290 microg/L), with 100% diagnostic sensitivity at the proposed Substance Abuse and Mental Health Services Administration screening cutoff of 4 microg/L cannabinoids and GC-MS cutoff of 2 microg/L THC. Forty-seven specimens screened positive but were not confirmed by 2D-GC-MS, yielding 89.5% diagnostic specificity and 90.6% diagnostic efficiency. Thirty-one of 47 unconfirmed immunoassay positive specimens were from 1 individual and contained >400 ng/L THCCOOH, potentially contributing to cross-reactivity. CONCLUSIONS The Immunalysis Sweat/OF THC Direct ELISA is an effective screening procedure for detecting cannabinoids in OF.


Clinical Chemistry | 2013

Oral Fluid Cannabinoids in Chronic Cannabis Smokers during Oral Δ9-Tetrahydrocannabinol Therapy and Smoked Cannabis Challenge

Dayong Lee; Ryan Vandrey; Damodara R. Mendu; Sebastien Anizan; Garry Milman; Jeannie A. Murray; Allan J. Barnes; Marilyn A. Huestis

BACKGROUND Oral Δ(9)-tetrahydrocannabinol (THC) is effective for attenuating cannabis withdrawal and may benefit treatment of cannabis use disorders. Oral fluid (OF) cannabinoid testing, increasing in forensic and workplace settings, could be valuable for monitoring during cannabis treatment. METHODS Eleven cannabis smokers resided on a closed research unit for 51 days and received daily 0, 30, 60, and 120 mg of oral THC in divided doses for 5 days. There was a 5-puff smoked cannabis challenge on the fifth day. Each medication session was separated by 9 days of ad libitum cannabis smoking. OF was collected the evening before and throughout oral THC sessions and analyzed by 2-dimensional GC-MS for THC, cannabidiol (CBD), cannabinol (CBN), 11-hydroxy-THC (11-OH-THC), and 11-nor-9-carboxy-THC (THCCOOH). RESULTS During all oral THC administrations, THC OF concentrations decreased to ≤ 78.2, 33.2, and 1.4 μg/L by 24, 48, and 72 h, respectively. CBN also decreased over time, with concentrations 10-fold lower than THC, with none detected beyond 69 h. CBD and 11-OH-THC were rarely detected, only within 19 and 1.6 h after smoking, respectively. THCCOOH OF concentrations were dose dependent and increased over time during 120-mg THC dosing. After cannabis smoking, THC, CBN, and THCCOOH concentrations showed a significant dose effect and decreased significantly over time. CONCLUSIONS Oral THC dosing significantly affected OF THCCOOH but minimally contributed to THC OF concentrations; prior ad libitum smoking was the primary source of THC, CBD, and CBN. Higher cannabinoid concentrations following active oral THC administrations vs placebo suggest a compensatory effect of THC tolerance on smoking topography.


Journal of Analytical Toxicology | 2009

Urinary MDMA, MDA, HMMA, and HMA Excretion Following Controlled MDMA Administration to Humans

Tsadik T. Abraham; Allan J. Barnes; Ross H. Lowe; Erin A. Kolbrich Spargo; Garry Milman; Stéphane Pirnay; David A. Gorelick; Robert S. Goodwin; Marilyn A. Huestis

Collaboration


Dive into the Garry Milman's collaboration.

Top Co-Authors

Avatar

Marilyn A. Huestis

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Allan J. Barnes

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar

Dayong Lee

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David M. Schwope

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar

Robert S. Goodwin

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Damodara R. Mendu

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar

Eugene W. Schwilke

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar

Ryan Vandrey

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge