You are on page 1of 8

Neuropharmacology 170 (2020) 107788

Contents lists available at ScienceDirect

Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm

Endocrine effects of the novel ghrelin receptor inverse agonist PF-5190457: T


Results from a placebo-controlled human laboratory alcohol co-
administration study in heavy drinkers
Mary R. Leea, Mehdi Farokhniaa,b, Enoch Cobbinac, Anitha Saravanakumarc, Xiaobai Lid,
Jillian T. Battistaa, Lisa A. Farinellia, Fatemeh Akhlaghic, Lorenzo Leggioa,b,e,f,∗
a
Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and
Biological Research and National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
b
Center on Compulsive Behaviors, National Institutes of Health, Bethesda, MD, USA
c
Clinical Pharmacokinetics Research Laboratory, Department of Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI,
USA
d
Biostatistics and Clinical Epidemiology Service, Clinical Center, National Institutes of Health, Bethesda, MD, USA
e
Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
f
Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA

HIGHLIGHTS

• We tested PF-5190457, a ghrelin receptor inverse agonist, in heavy drinkers.


• We assessed the effect of PF-5190457 on neuroendocrine hormones.
• Blood hormone levels were measured during dosing phase and alcohol administration.
• Neuroendocrine hormones remain largely unaffected by PF-5190457.

ARTICLE INFO ABSTRACT

Keywords: Both animal and human work suggests that the ghrelin system may be involved in the mechanisms that regulate
Ghrelin the development and maintenance of alcohol use disorder. Previously, in a Phase 1b study, we tested phar-
GHS-R1a macological blockade of the growth hormone secretagogue receptor 1a (GHS-R1a, also known as the ghrelin
PF-5190457 receptor), in heavy drinking individuals with PF-5190457, an orally bioavailable, potent and selective GHS-R1a
Alcohol
inverse agonist. We report here the effects of PF-5190457 on endocrine blood concentrations of amylin, gastric
Heavy drinking
inhibitory polypeptide, glucagon-like peptide 1, insulin, leptin, pancreatic polypeptide, peptide YY, thyroid
Alcohol use disorder
Hormones stimulating hormone, free triiodothyronine (T3), thyroxine (T4), cortisol, prolactin, and glucose during PF-
Neuroendocrinology 5190457 dosing, as compared to placebo, in absence of alcohol as well as during an alcohol challenge when PF-
5190457 was on steady-state. Blood hormone levels were largely unaffected by PF-5190457, both during dosing
and in the context of alcohol challenge. The safety-related relevance of these findings to further develop PF-
5190547 in alcohol use disorder is discussed.
Clinicaltrials.gov: NCT02039349.
This article is part of the special issue on ‘Neuropeptides’.

1. Introduction was originally discovered for its function in regulating growth hormone
(GH) secretion, subsequent work identified ghrelin as a hormone with
The peptide ghrelin is a gut hormone produced by endocrine cells pleiotropic functions that include (but are not limited to) regulation of
mainly localized in the stomach (Kojima et al., 1999). While ghrelin appetite, food intake, energy expenditure, and glucose homeostasis


Corresponding author. Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology NIAAA & NIDA, NIH; 10 Center Drive (10CRC/15330) MSC
1108, Room 1-5429 Bethesda, MD, 20892-1108, USA.
E-mail address: lorenzo.leggio@nih.gov (L. Leggio).

https://doi.org/10.1016/j.neuropharm.2019.107788
Received 18 April 2019; Received in revised form 30 August 2019; Accepted 19 September 2019
Available online 23 September 2019
0028-3908/ Published by Elsevier Ltd.
M.R. Lee, et al. Neuropharmacology 170 (2020) 107788

(Leggio, 2010; Muller et al., 2015). Ghrelin is acylated by the ghrelin-O- late increase in IGF-1 concentrations under both drug conditions (50 mg
acyltransferase (GOAT) enzyme to acyl-ghrelin (hereafter mostly re- b.i.d. and 100 mg b.i.d.) compared to placebo, suggesting that IGF-1
ferred to as ‘ghrelin’), which then binds to its G-protein coupled GH might represent a biomarker of the downstream effects of GHS-R1a
secretagogue receptor (GHS-R1a) to execute its physiological functions blockade with PF-5190457 in heavy drinkers (Lee et al., 2018). Overall,
(Howard et al., 1996). these results indicated the safety of PF-5190457, alone and when co-
Both animal and human work suggests that the ghrelin system may administered with alcohol. In summary, PF-5190457 affects ghrelin
be involved in the mechanisms that regulate the development and signaling, but in a complex way that may become more intricate with
maintenance of alcohol use disorder (AUD). As recently reviewed co-administration of alcohol.
(Farokhnia et al., 2019; Jerlhag, 2019; Morris et al., 2018; Zallar et al., We report here the effects of PF-5190457 on a wide range of other
2017), results from experiments conducted by independent laboratories endocrine outcomes investigated in this study (Lee et al., 2018). Spe-
and using different rodent models indicate that ghrelin administration cifically, given the effect of ghrelin to inhibit glucose-stimulated insulin
leads to an increase in alcohol-related outcomes, such as alcohol con- secretion and to impair glucose tolerance, we measured blood insulin
ditioned place preference (a proxy of reward), as well as alcohol intake, and glucose concentrations, as well as other hormones with known
preference, and self-administration. These effects are attenuated by effects on glucose homeostasis, appetite, and food intake. These hor-
transgenic knock-out or pharmacological blockage (D-Lys3-GHRP-6, mones include amylin, gastric inhibitory polypeptide (GIP), glucagon-
BIM28163, or JMV2959) of the ghrelin receptor. In addition, human like peptide 1 (GLP-1), insulin, leptin, pancreatic polypeptide (PP),
studies have reported a relationship between endogenous ghrelin con- peptide YY (PYY), thyroid stimulating hormone (TSH), free triio-
centrations and alcohol craving, risk of relapse, subjective responses to dothyronine (T3), and thyroxine (T4). Finally, given the role of ghrelin
alcohol, and brain activity in response to alcohol cues, suggesting that in stress regulation, as well as the emerging complex interactions be-
endogenous ghrelin signaling is positively associated with alcohol-re- tween ghrelin, stress pathways, and AUD (for review, see (Morris et al.,
lated behaviors (for reviews, see (Farokhnia et al., 2019; Jerlhag, 2019; 2018)), we also measured blood concentrations of the stress hormones
Morris et al., 2018; Zallar et al., 2017)). cortisol and prolactin.
Further human studies support a role of the ghrelin system in AUD The endocrine-focused analyses presented here are important at
by showing that manipulations of the ghrelin system lead to changes in several levels, as they may contribute to a better understanding of: 1)
alcohol-related outcomes. Specifically, two double-blind, placebo-con- the safety of PF-5190457 administration, with respect to its effect on
trolled studies demonstrated that intravenous administration of ghrelin these endocrine pathways, when administered alone or co-administered
increases cue-induced alcohol craving in a bar-like setting (Leggio et al., with alcohol; 2) the potential indirect endocrine effects of GHS-R1a
2014) and alcohol self-administration using a progressive ratio alcohol blockade via PF-5190457 that may contribute to the downstream me-
intravenous paradigm (Farokhnia et al., 2018). In another experimental chanisms of action of this novel compound; and 3) the potential for
human study, forced water intake led to reduction in endogenous using specific hormones as peripheral endocrine biomarkers of PF-
ghrelin concentrations (possibly due to the stomach distension), which 5190457's activity in heavy alcohol drinkers and in the context of al-
in turn was associated with reduced alcohol craving (Koopmann et al., cohol use.
2017).
Based on the rodent and human literature summarized above, we 2. Materials and methods
tested the effects of GHS-R1a pharmacological blockade in heavy
drinking individuals. PF-5190457 is an orally bioavailable, potent, and 2.1. Study design
selective GHS-R1a inverse agonist (Bhattacharya et al., 2014) that in-
hibits the receptor's constitutive activity and competitively blocks its This was a Phase 1b, within-subject, dose-escalating, single-blind,
activation by acyl-ghrelin (Kong et al., 2016). PF-5190457 is the first placebo-controlled human laboratory study with three oral drug con-
GHS-R1a inverse agonist that progressed to a Phase 1a clinical trial, ditions: placebo b.i.d., PF-5190457 50 mg b.i.d., and 100 mg b.i.d.,
conducted by the manufacturer, where PF-5190457 was well-tolerated conducted at the NIH Clinical Center, Bethesda, Maryland, USA.
and safe in healthy volunteers (Denney et al., 2017). We also conducted
a translational study starting with a set of rat experiments to rule out 2.2. Study approvals
significant safety concerns due to potential interactions between PF-
5190457 and alcohol (Lee et al., 2018). Next, in heavy drinking in- This human study was approved by the NIH Addictions Institutional
dividuals (Phase 1b human laboratory study), we examined the safety Review Board and monitored by a Data Safety Monitoring Board.
of PF-5190457, alone and when administered with alcohol (Lee et al., Written informed consent was obtained from participants prior to in-
2018). clusion in the study. The study was conducted under a federal
In the Phase 1b human laboratory study, PF-5190457 or placebo Certificate of Confidentiality. The use of PF-5190457 in this study was
was administered up to steady-state (dosing phase), followed by oral under the Food and Drug Administration Investigational New Drug
alcohol administration (alcohol challenge phase). We determined that 119 365. Alcohol administration procedures were consistent with the
PF-5190457 (50 mg b.i.d. and 100 mg b.i.d.) alone and co-administered NIAAA Council Guidelines on Alcohol Administration: (https://www.
with alcohol was safe and well tolerated. We also reported that there niaaa.nih.gov/Resources/ResearchResources/job22.htm).
were no pharmacokinetic (PK) changes in either PF-5190457 or alcohol
during their co-administration. Additionally, preliminary data in- 2.3. Study sample
dicated that PF-5190457 may reduce cue-induced craving for alcohol
and food in a bar-like setting (Lee et al., 2018). 1. Eligible participants were male and female active, heavy alcohol
Furthermore, we investigated the potential effects of PF-5190457 on drinkers (≥21 drinks/week for men or ≥ 14 drinks/week for
endocrine pathways related to the ghrelin system, either directly (acyl- women, based on the 90-day timeline follow-back at screening) and
ghrelin, total ghrelin, acyl-to-total ghrelin ratio) or indirectly (GH, in- were not seeking treatment for AUD. Recruitment took place via
sulin-like growth factor 1 (IGF-1), the latter being a clinically used word of mouth, as well as local and online advertisements. After a
surrogate measure of GH (Lee et al., 2018). We found an increase in preliminary phone pre-screening, potentially eligible participants
total ghrelin levels in the PF-5190457 100 mg condition, but limited to came to NIH and signed an informed consent to enroll in a screening
the alcohol challenge phase, while during the dosing phase there was a protocol for determination of suitability for enrollment in the clin-
significant effect of PF-5190457 100 mg to reduce the acyl-to-total ical study with PF-5190457. A full list of the inclusion and exclusion
ghrelin ratio. Additionally, during the alcohol challenge, there was a criteria is reported in the Supplementary Appendix 1.

2
M.R. Lee, et al. Neuropharmacology 170 (2020) 107788

2. Fourteen eligible participants signed the informed consent and administration. Then, alcohol was administered 30 min after study drug
twelve completed the study. Demographics and baseline character- administration and blood samples were taken at: +30, +60, +120,
istics are reported in the supplementary material of the main paper +180, +240, +360, +480, +1350 and + 1440. Day 3 glucose
(Lee et al., 2018) and here in the Supplementary Table S1. In ad- monitoring was done at −30, +30, +60, +120, +180, +240, +360,
dition to being heavy drinkers, all but one participant had a current +480, +1350, +1440.
diagnosis of alcohol dependence based on the Diagnostic and Sta-
tistical Manual of Mental Disorders IV Edition – Text Revision (DSM- 2.4.3. Standardization and timing of meals
IV-TR) criteria. During each inpatient stay, participants received meals standardized
for macronutrients and caloric content. For additional details, please
2.4. Study procedures see (Lee et al., 2018). The timing of the meals relative to PF-5190457
dosing was as follows: breakfast was at −120, before daily PF-5190457
After signing the study-specific informed consent, research proce- dosing; Lunch was eaten at +210, and dinner at +570.
dures started. There were three identical inpatient visits (Visits 1–3)
separated by at least three days. The inpatient setting ensured full 2.5. Assays for blood hormone assessments
compliance with the study medication, procedures, and alcohol ab-
stinence, and permitted strict safety monitoring. At each of the three The research plasma samples were used to measure active amylin
visits, PF-5190457/placebo administration took place twice on Day 1, (here referred to as amylin), GIP, active GLP-1 (here referred to as GLP-
twice on Day 2, and on the morning of Day 3 (dosing phase). On Day 3, 1), insulin, leptin, PP and PYY. These plasma samples derived from
approximately 30 min after the administration of PF-5190457, a drink blood tubes centrifuged within 30 min post-collection (relative cen-
containing the same type of alcohol (40% Alcohol by Volume; 80% trifugal force: 1700×g, temperature: 4 °C, centrifugation time: 15 min),
Proof) for all subjects was administered (alcohol challenge phase). The and pre-treated with 4-(2-aminoethyl)benzenesulfonyl fluoride hydro-
quantity of alcohol was calculated based on total body water for each chloride (Roche Diagnostics GmbH, Germany – Pefabloc® SC), di-
subject to achieve a blood alcohol concentration of approximately peptidyl peptidase IV inhibitor (EMD Millipore Corp., Billerica, MA –
0.06 mg% (Watson, 1989). At each of the three visits, participants were Cat. #DPP4-010), and a protease inhibitor cocktail (Sigma-Aldrich Inc.,
discharged on Day 4 after a clinical assessment. Approximately 1–2 Saint Louis, MO – Cat. #P8340) prior to blood collection. Samples were
weeks after Visit 3, a brief outpatient clinical assessment took place, inverted 10 times and kept on ice after collection. These hormones were
during which participants’ health, safety, and alcohol consumption measured using a Millipore Human Metabolic Hormone Magnetic Bead
were evaluated and counseling was delivered per NIAAA guidelines: Panel 96-Well Plate MILLIPLEX® MAP kit (EMD Millipore Corp.,
(https://pubs.niaaa.nih.gov/publications/Practitioner/ Billerica, MA – Cat. #HMHEMAG-34K). The assay was performed on
CliniciansGuide2005/clinicians_guide.htm). Participants received fluorescence-coded magnetic beads coated with capture antibodies
monetary compensation for participation in this study. specific for each marker. Introduction of biotinylated detection anti-
The study timeline is outlined in the supplementary material of the body and streptavidin-phycoerythrin permitted simultaneous detection
main paper (Lee et al., 2018) and here in the Supplementary Tables of all analytes on the MAGPIX® instrument (Luminex Corp., Austin, TX).
S2–S3. These multiplex data were pre-processed and analyzed in the MILLIP-
Additional details, procedures, and assessments, not relevant to the LEX® Analyst software (Version 3.5 – EMD Millipore Corp., Billerica,
analysis reported here, are described in the main paper (Lee et al., MA) to calculate the concentration of each hormone. Capillary glucose
2018). concentrations were assessed with a StatStrip Glucose Meter, in con-
junction with StatStrip Xpress Test Strips (Nova Biomedical), using the
2.4.1. Timepoints for blood collection during the dosing phase Point of Care Testing nursing procedures at the NIH Clinical Center.
At each of the three visits, during the PF-5190457/placebo dosing The thyroid panel (TSH, free T3, and thyroxine) and the stress
phase (Days 1–3), research blood samples were collected and processed, hormones, cortisol and prolactin, were assayed at the NIH Clinical
and plasma samples were stored at −80 °C for future analyses. Center Department of Laboratory Medicine. Electrochemiluminescence
Specifically, these research blood samples were collected at six time- immunoassays were run on a cobas® 600 analyzer (Roche Diagnostics
points in the morning of each day, Day 1–3: 90 min before the 1st dosing International Ltd., Switzerland) to measure TSH, free T3, and thyroxine
(T1), 75 min after the 1st dosing (T2), 90 min before the 3rd dosing concentrations. Chemiluminescence immunoassays were run on an
(T3), 75 min after the 3rd dosing (T4), 60 min before the 5th dosing IMMULITE® 2000 XPi (Siemens Healthcare Diagnostics Inc., Tarrytown,
(T5) and 15 min after the 5th dosing (T6; before the alcohol adminis- NY) to measure cortisol and prolactin.
tration began). The differences in the timing of blood samples collection
reflect the fact that the T6 blood draw (+15 min after the fifth dosing) 2.6. Statistical analysis
was drawn on the morning of the alcohol challenge. The alcohol chal-
lenge began 30 min after study drug dosing, therefore, required that the The serial measures of blood hormone levels from the dosing phase
T6 blood draw to be 15 min after dosing to acquire another morning and the alcohol challenge phase were analyzed separately. Mean and
blood sample before alcohol administration. standard deviation for each hormone outcome was reported at each
Capillary glucose concentrations were also assessed with finger- assessment timepoint for each DOSE condition (placebo, PF-5190457
sticks at the four timepoints during Day 1 and Day 2, specifically at 50 mg b.i.d., and 100 mg b.i.d.). To assess the treatment effect in these
−30, +30, +690, and +750. Furthermore, blood samples for clinical hormones, data were analyzed using linear mixed effects models using
labs (including hormones as detailed below) were transferred to the DOSE, TIME, and TIME × DOSE interaction as the fixed effects. Within-
NIH Clinical Center Department of Laboratory Medicine, immediately subject correlation among the repeated measures were taken into ac-
after collection, and processed and analyzed there, as part of the pro- count by the subject random effect. For the alcohol challenge phase, the
tocol clinical assessments at three timepoints (T1, T3, and T5). last two timepoints were collected the morning after (Day 4) and there
was a 14-h gap between the last Day 3 blood draw (7th) and the last
2.4.2. Timepoints for blood collection during the alcohol challenge phase two (8th and 9th) timepoints (Day 4). Therefore, we did not include the
At each of the three visits, during the alcohol challenge phase (Day last two timepoints in the main analyses in order to maintain relatively
3), research blood samples were collected from a peripheral vein, equal time intervals between the data points. That is, without including
processed and stored at −80 °C in our laboratory for future analyses. the remote two timepoints, we avoided making extrapolations for that
Blood was drawn 60 min before and 15 min after study drug 14-h gap. However, the 8th and 9th timepoints are still represented in

3
M.R. Lee, et al. Neuropharmacology 170 (2020) 107788

Table 1
Main effect of TIME, DOSE, and TIME × DOSE interaction on blood hormones
and glucose concentrations during the dosing phase.
Variable Main effect INTERACTION Transformation
TIME X DOSE
TIME DOSE

Amylin <0.001 0.008 0.866 Log-transformed


GIP <0.001 0.970 0.820 Square root transformed
GLP-1 0.013 0.152 0.460 Square root transformed
Insulin <0.001 <0.001 0.220 Square root transformed
Leptin <0.001 0.780 0.970 Log-transformed
PP 0.16 0.270 0.400 Square root transformed
PYY 0.048 0.122 0.140 Square root transformed
TSH <0.015 0.011 0.051 Square root transformed
free T3 0.990 0.930 0.630 None
Thyroxine 0.040 0.860 0.320 None
Cortisol <0.001 0.640 0.450 None
Prolactin 0.040 0.490 0.850 None
Glucose <0.001 0.130 0.990 Square root transformed
Fig. 1. Blood concentration [Mean (±SD)] of thyroid stimulating hormone
(TSH) at 90 min before (timepoints 1,3,5) drug dosing on Day 1,2,3. Drug doses:
Placebo, PF-5190457, 50 mg and 100 mg.
Table 2
Main effect of TIME, DOSE, and TIME × DOSE interaction on blood hormones
and glucose concentrations during the alcohol challenge phase. doses of PF-5190457, TSH levels remained similar to baseline (Fig. 1).
Variable Main Effect INTERACTION Transformation This finding was observed in the absence of a main effect of DOSE or
TIME X DOSE TIME × DOSE interaction for free T3 or thyroxine. There was a sig-
TIME DOSE nificant main effect of DOSE, as well for TSH (p = 0.011). There were
also significant main effects of DOSE for amylin (p = 0.008) and insulin
Amylin 0.034 0.030 0.950 Log-transformed
GIP <0.001 0.040 0.300 Square root transformed (p < 0.001), so that there were lower concentrations of insulin (Fig. 2)
GLP-1 <0.001 0.560 0.710 Square root transformed and amylin (Fig. 3) under PF-5190457, as compared to placebo. There
Insulin <0.001 <0.001 0.073 Square root transformed was a significant main effect of TIME for all but 2 of the hormones, as
Leptin <0.001 <0.001 0.720 Log-transformed detailed in Table 1.
PP <0.001 0.220 0.460 Square root transformed
PYY 0.330 0.160 0.590 Square root transformed
The supplementary figures outline the non-significant results related
Glucose <0.001 0.170 0.100 Square root transformed to the lack of a main effect of DOSE for blood concentrations of GIP
(Fig. S1A), GLP-1 (Fig. S1B), leptin (Fig. S1C), PP (Fig. S1D), PYY (Fig.
S1E), free T3 (Fig. S1F), thyroxine (Fig. S1G), cortisol (Fig. S1H), pro-
the figures for descriptive purposes and to allow for a visual inspection. lactin (Fig. S1I), and glucose (Fig. S1J).
The logarithm or square root transformation was applied to the Finally, the heatmap (amylin, GIP, GLP-1, insulin, leptin, PP, and
outcome variables when the model normality assumption was violated PYY only) and the related dendogram in Fig. 4 showed a similar effect
(see also Tables 1–2). Values below the lower limit of quantitation for for leptin, amylin, and insulin, where AUC decreased in the PF-
each assay were set to 1/2 of the lower limit of quantitation (Keizer 5190457 100 mg b.i.d. condition compared to the placebo b.i.d. con-
et al., 2015). A p value of 0.05 or lower was considered statistically dition. Conversely, AUC for PP, GLP, PYY, and GIP in the PF-
significant. All statistical analyses were conducted in SAS (version 9.4, 5190457 100 mg b.i.d. condition was greater than the placebo condi-
SAS Institute Inc., Cary, NC). tion.
Finally, we conducted a heatmap analysis which was limited to the
hormones for which six timepoints were available during the dosing
phase (amylin, GIP, GLP-1, insulin, leptin, PP, and PYY only). We did
not include hormone measurements during the alcohol challenge on
Day 3 (due to the potential confounding effect of alcohol on these
measures) and we examined only the placebo and the highest dose of
PF-5190457 (100 mg b.i.d.). Specifically, the aggregate drug effect was
summarized by the area under the hormone-concentration-time curve
(AUC) for the PF-5190457 100 mg b.i.d. and placebo b.i.d. conditions.
The difference in the log-transformed AUCs between the two conditions
was calculated for each subject. We used a heatmap analysis to examine
whether the effect of PF-5190457 dosing compared to placebo was si-
milar amongst any of the hormones, with respect to the direction and
magnitude of the effect. The heatmap clustering analysis was conducted
using the heatmap package with RStudio (http://www.rstudio.org/,
integrated development environment (IDE) for R, Boston, MA, USA).

3. Results

3.1. Dosing phase

Results (p values) are outlined in Table 1. TSH was the only hor- Fig. 2. Blood concentration [Mean (±SD)] of insulin at 90 min before (time-
mone with a nominal trend TIME × DOSE interaction (p = 0.051). In points 1,3,5) and 75 min after (timepoints 2,4,6) drug dosing on Day 1,2,3.
the placebo condition, TSH decreased over time, while under the 2 Drug doses: Placebo, PF-5190457, 50 mg and 100 mg.

4
M.R. Lee, et al. Neuropharmacology 170 (2020) 107788

4. Discussion

This report provides additional information on the effect of a GHS-


R1a inverse agonist (PF-5190457) (Lee et al., 2018) on blood con-
centrations of endocrine hormones when PF-5190457 is dosed to steady
state and administered with alcohol. The study population comprised of
individuals who were heavy drinkers and mostly (11 out of 12) with a
DSM-IV-TR diagnosis of alcohol dependence. Notwithstanding the ex-
ploratory and non-comprehensive nature of this work, the present
secondary analysis represented an opportunity to expand our knowl-
edge on the safety and endocrine effects of GHS-R1a blockade via PF-
5190457, in heavy drinkers and during an acute alcohol administration.
There was evidence, as we previously reported (Lee et al., 2018),
that PF-5190457 modulated endogenous ghrelin levels, as there was a
dose-dependent reduction in the acyl-to-total ghrelin ratio. Aside from
this finding, the majority of the hormones reported here were not af-
fected by PF-5190457, either during the dosing phase or during the
alcohol challenge phase. The Phase 1a first-in-human study in healthy
Fig. 3. Blood concentration [Mean (±SD)] of amylin at 90 min before (time- controls also reported no significant effects of PF-5190457 on blood
points 1,3,5) and 75 min after (timepoints 2,4,6) drug dosing on Day 1,2,3. levels of a partially overlapping list of neurotransmitters and hormones:
Drug doses: Placebo, PF-5190457, 50 mg and 100 mg. dopamine, noradrenaline, adrenaline, glucagon, C-peptide, insulin, and
PP (Denney et al., 2017). A comprehensive, side-by-side comparison of
the two studies is not possible, given differences in study design, times
of blood sample collection, and fasting conditions.
Most importantly, unlike the previous Phase 1a study in healthy
volunteers (Denney et al., 2017), the present Phase 1b study was con-
ducted in heavy drinkers, most of whom (11 out of 12) were alcohol
dependent (DSM-IV-TR) and included an alcohol administration ex-
periment. AUD may be associated with several changes in a variety of
endocrine systems (for a review, see (Kenna et al., 2012)). As such,
assessing the safety of PF-5190457 administration in heavy drinkers in
terms of endocrine endpoints, including when co-administered with
alcohol, offers highly relevant information given the role of PF-
5190457 as a new compound currently under study to investigate GHS-
R1a inverse agonism as a novel pharmacotherapy for AUD patients.
Indeed, the lack of clinically-relevant safety concerns when PF-5190457
was co-administered with alcohol is critical, given the importance of
ruling out clinically-relevant drug-alcohol interactions early on, from a
medication development perspective (Haass-Koffler et al., 2017).
The significant TIME × DOSE interaction indicates that PF-5190457
affected the time course of blood TSH levels over the 3 days of study.
Fig. 4. Heatmap representation of the difference in the hormone-concentration- Specifically, in the placebo condition, TSH levels fell over the 3 days.
time curve (AUC) encompassing Days 1,2,3 for the PF-5190457 100 mg minus This decrease would be expected in this largely alcohol dependent
the placebo conditions for each of the 12 subjects who completed the study population, who were current heavy drinkers. Previous studies have
(N101-N114). The heatmap analysis was limited to the hormones for which six
reported that individuals with AUD have elevated TSH levels compared
timepoints were available during the dosing phase: amylin, gastric inhibitory
to controls, and that during acute abstinence, TSH levels fall. Under PF-
polypeptide (GIP), glucagon-like peptide 1 (GLP-1), insulin, leptin, pancreatic
5190457, this decrease was attenuated. Human studies administering
polypeptide (PP), and peptide YY.
ghrelin reported a decrease in TSH and an increase in T4 (Kluge et al.,
2010). Accordingly, an inverse agonist theoretically could increase
3.2. Alcohol challenge phase TSH, however we found that this biochemical increase in TSH was not
accompanied by any effect of PF-5190457 on either free T3 or T4. As
Results (p values) are outlined in Table 2. Insulin was the only such, while it is unlikely that PF-5190457 may affect the thyroid
hormone with a trend significant TIME × DOSE interaction function, these results suggest that TSH might represent a potential
(p = 0.073), as well as a significant main effect (p < 0.001), where PF- marker of PF-5190457's activity in this population of heavy drinkers.
5190457, co-administered with alcohol, reduced blood insulin levels Furthermore, as outlined in Fig. 1, it is possible that the effect of PF-
(Fig. 5a). There was a significant main effect of TIME for all variables 5190457 on TSH here described is apparent only at the highest dose
except PYY, as detailed in Table 2. (100 mg).
There was a significant main effect of DOSE for amylin (p = 0.03), It has been shown that central administration of TSH reduces food,
GIP (p = 0.04), and leptin (p < 0.001), with higher concentrations of but not water, intake in rats (Lin et al., 1983), suggesting that TSH may
amylin (Fig. 5b) and lower concentrations of GIP (Fig. 5c) and leptin act through a central mechanism to influence physiological or beha-
(Fig. 5d) under PF-5190457, co-administered with alcohol. The sup- vioral functions like appetite and food intake (Amin et al., 2011; Lin
plementary material includes the figures of the non-significant results et al., 1983). In the parent paper (Lee et al., 2018), we reported that, on
related to the lack of a main effect of DOSE for blood concentrations of Day 2 (dosing phase), PF-5190457 versus placebo reduced both al-
GLP-1 (Fig. S1B), PP (Fig. S1D), PYY (Fig. S1E) and glucose (Fig. S1J). cohol- and food cue-induced cravings. Albeit speculative, it is possible
to hypothesize that TSH signaling may centrally contribute to the ef-
fects of PF-5190457 on alcohol- and food-seeking behaviors, without

5
M.R. Lee, et al. Neuropharmacology 170 (2020) 107788

Fig. 5. Blood concentrations [Mean (±SD)] of A) Insulin, B) Amylin, C) Gastrointestinal Peptide (GIP), and D) Leptin during the alcohol challenge, which was
performed on Day 3. Alcohol was administered at +30, lunch at +210, and dinner at +570. Timepoints +1350 and + 1440 were not included in the analysis and
their inclusion in the figures is only for illustrative purposes.

affecting free T3 and thyroxine, and their peripheral effects on energy and amylin levels, as they were not accompanied by changes in glucose
expenditure and/or metabolism. Notably, while the role of the hy- levels, therefore it is unlikely that PF-5190457 affects peripheral glu-
pothalamic-pituitary-thyroid axis on appetite is well-known, previous cose homeostasis. In other words, while the present findings and pre-
work from our group indicated a negative relationship between blood vious work (Broglio et al., 2001; Haass-Koffler et al., 2016; Tong et al.,
TSH levels and alcohol craving (Aoun et al., 2015; Leggio et al., 2008), 2010) support a link between the ghrelin system and insulin signaling
and higher TSH levels in AUD patients compared to controls (Lee et al., in humans, the effect of the inverse agonist on insulin levels is the
2015). Although preliminary and speculative, the latter previous find- opposite from what would be expected given the effect of ghrelin, itself,
ings fit well with the present data, where PF-5190457 increased TSH to reduce insulin levels. This inconsistency most likely reflects: 1) the
levels (present results) while it reduced alcohol craving (reported pre- complexity of pharmacological blockade of GHS-R1a, whose down-
viously in (Lee et al., 2018)). stream effects on insulin signaling may considerably vary and 2) the
PF-5190457 significantly reduced blood concentrations of glucose- difficulty of comparing the dosing phase and the alcohol challenge
regulating hormones, chiefly insulin and amylin, during the dosing phase within our own present study, given the different experimental
phase. This result was also apparent in the heatmap analysis, where the conditions (e.g., timepoints and caloric control) applied.
AUC variable, effectively a time average for these two hormones, was in The results discussed above are important and potentially inter-
the same cluster. During the alcohol challenge phase, insulin levels esting, considering they are drawn from humans and from a clinically-
were higher in the placebo condition both before and after lunch (lunch relevant sample. However, the considerations above remain speculative
was at timepoint 210). The reason for the late (timepoint 480) rebound and bed-to-bench approaches are needed to examine mechanisms un-
in insulin levels, which drives the trend-level DOSE × TIME interaction, derlying the effect of PF-5190457 on hormones like TSH, insulin,
is unclear. This rebound was also seen in amylin, although at an earlier amylin, and probably others.
timepoint, but there was no significant interaction. It is also important We speculate that the increase in TSH, in the absence of changes in
to keep in mind the expected variability in hormone concentrations free T3 and thyroxine levels, as well as the reduction in insulin and
across individuals, which may limit the interpretation of the results. For amylin levels, in the absence of effects on blood glucose levels, suggest
example, although the small sample does not allow for additional that PF-5190457 may not result in total or continuous blockade at GHS-
analysis of sub-groups, it is worth noticing that a visual inspection of R1a. The previous Phase 1a first-in-human study was terminated due to
the heatmap dendrogram suggests that the decreasing trend of leptin, tachyphylaxis, which was limited to peripheral, but not central effects
amylin and insulin concentrations observed in the last 7 individuals is (Denney et al., 2017). Therefore, there is the possibility that the effects
not exhibited by the first 5 individuals. reported here on TSH and insulin, which do not result in downstream
The results presented here represent biochemical changes in insulin metabolic effects, are a consequence of central blockade of GHS-R1a,

6
M.R. Lee, et al. Neuropharmacology 170 (2020) 107788

which may be incomplete; that is, PF-5190457 may cause continuous Acknowledgments
blockade at the peripheral GHS-R1a, whereas central blockade is not
continuous, allowing for enduring efficacy (Denney et al., 2017). In- The authors would like to thank the clinical and research staff in-
deed, our rat experiments indicate some PF-5190457 brain penetrance; volved in data collection and support at the National Institute on
however, this work was limited to measuring PF-5190457 concentra- Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical
tions in brain homogenates and we did not use any isotopically labeled and Biological Research, i.e. in the NIAAA/NIDA Section on Clinical
PF-5190457. Finally, both the present results and the previous Phase 1a Psychoneuroendocrinology and Neuropsychopharmacology and in the
study (Denney et al., 2017) did not find an effect of PF-5190457 on NIAAA clinical intramural program. The authors would also like to
blood PP levels, which is largely under vagal control (Schwartz, 1983) thank the research staff involved in samples and data analysis in the
and is considered a marker of peripheral vagus nerve activation Clinical Pharmacokinetics Research Laboratory at the University of
(Tamboli et al., 2017). Rhode Island. The authors would like to thank the clinical and research
PF-5190457 is a novel compound and the information provided staff involved in data collection and patient care at the NIH Clinical
here is the first of its kind, given the specific population we studied and Center, i.e. in the Department of Nursing (the nurses of the 1SE
the experimental design that included alcohol administration. Inpatient Unit and of the 1-HALC 1SE Outpatient Clinic), in the
Furthermore, the study was conducted in a well-controlled research Department of Nutrition, and in the Department of Pharmacy.
inpatient setting, which strengthened compliance, consistency across Furthermore, the authors would like to express their gratitude to the
visits, and safety monitoring. However, this work also has important participants who took part in this study. Finally, the authors would like
limitations. First, the sample size was small (N = 12 completers), al- to thank the Steering Committee of the UH2/UH3-TR000963 grant (PIs:
though the within-subject design attenuated this limitation. Some of the Drs Lorenzo Leggio and Fatemeh Akhlaghi) whose members included
hormone results (TSH, free T3, thyroxine, cortisol, and prolactin) were members from the NIAAA Division of Medication Development (in
available for the dosing phase, but not for the alcohol challenge phase; particular Dr. Joanne Fertig), the Drug Development Partnership
this is a weakness that limits the interpretation of the present findings Programs of the National Center for Advancing Translational Sciences
but was necessary due to the safety-related need to limit the amount of (NCATS) and Pfizer, which kindly provided the study drug under the
blood drawn in a clinical research protocol. We did not correct for NCATS grant UH2/UH3-TR000963. Pfizer did not have any role in the
multiple testing, an approach consistent with the exploratory nature of study design, execution, or interpretation of the results, and this pub-
these analyses. Therefore, caution needs to be exercised given the risk lication does not necessarily represent the official views of Pfizer.
for false positive results. Furthermore, the lack of an alcohol-matched
placebo (to make the design fully factorial) calls for future larger, Appendix A. Supplementary data
randomized studies that consider these limitations. Finally, it is im-
portant to note that, ~90% of study participants in our study were Supplementary data to this article can be found online at https://
African-Americans and, given the strict inclusion and exclusion criteria, doi.org/10.1016/j.neuropharm.2019.107788.
recruitment of females was very difficult (only one out of 12 com-
pleters). Therefore, these results may not be generalizable, and future References
studies are needed to further investigate PF-5190457 across individuals
with different race, as well as potential sex differences. Amin, A., Dhillo, W.S., Murphy, K.G., 2011. The central effects of thyroid hormones on
appetite. J. Thyroid Res. 2011, 306510.
Aoun, E.G., Lee, M.R., Haass-Koffler, C.L., Swift, R.M., Addolorato, G., Kenna, G.A.,
Leggio, L., 2015. Relationship between the thyroid axis and alcohol craving. Alcohol
Author contributions Alcohol 50, 24–29.
Bhattacharya, S.K., Andrews, K., Beveridge, R., Cameron, K.O., Chen, C., Dunn, M.,
Study neuroscientific basis, rationale, concept and design: LL; Provided Fernando, D., Gao, H., Hepworth, D., Jackson, V.M., Khot, V., Kong, J., Kosa, R.E.,
Lapham, K., Loria, P.M., Londregan, A.T., McClure, K.F., Orr, S.T., Patel, J., Rose, C.,
funding: LL, FA; Statistical analysis: XL; Acquisition and management of
Saenz, J., Stock, I.A., Storer, G., VanVolkenburg, M., Vrieze, D., Wang, G., Xiao, J.,
data: MRL, MF, EC, AK, JTB, LAF, FA, LL; Clinical and Safety monitoring: Zhang, Y., 2014. Discovery of PF-5190457, a potent, selective, and orally bioavail-
MRL, LL; Administrative, technical, or material support: MRL, MF, EC, AK, able ghrelin receptor inverse agonist clinical candidate. ACS Med. Chem. Lett. 5,
XL, JTB, LAF, FA, LL; Analysis and interpretation of data: MRL, MF, EC, 474–479.
Broglio, F., Arvat, E., Benso, A., Gottero, C., Muccioli, G., Papotti, M., van der Lely, A.J.,
AK, XL, JTB, LAF, FA, LL; Drafting the manuscript: MRL, LL. All authors Deghenghi, R., Ghigo, E., 2001. Ghrelin, a natural GH secretagogue produced by the
have critically reviewed the manuscript for important intellectual stomach, induces hyperglycemia and reduces insulin secretion in humans. J. Clin.
content and approved the final version of the manuscript. Endocrinol. Metab. 86, 5083–5086.
Denney, W.S., Sonnenberg, G.E., Carvajal-Gonzalez, S., Tuthill, T., Jackson, V.M., 2017.
Pharmacokinetics and pharmacodynamics of PF-05190457: the first oral ghrelin re-
ceptor inverse agonist to be profiled in healthy subjects. Br. J. Clin. Pharmacol. 83,
Funding source 326–338.
Farokhnia, M., Faulkner, M.L., Piacentino, D., Lee, M.R., Leggio, L., 2019. Ghrelin: from a
gut hormone to a potential therapeutic target for alcohol use disorder. Physiol.
This work was supported by NIH intramural funding ZIA-AA000218
Behav. 204, 49–57.
(Section on Clinical Psychoneuroendocrinology and Farokhnia, M., Grodin, E.N., Lee, M.R., Oot, E.N., Blackburn, A.N., Stangl, B.L., Schwandt,
Neuropsychopharmacology; PI: Dr. Lorenzo Leggio), jointly supported by M.L., Farinelli, L.A., Momenan, R., Ramchandani, V.A., Leggio, L., 2018. Exogenous
the Division of Intramural Clinical and Biological Research of the ghrelin administration increases alcohol self-administration and modulates brain
functional activity in heavy-drinking alcohol-dependent individuals. Mol. Psychiatry
National Institute on Alcohol Abuse and Alcoholism (NIAAA) and the 23, 2029–2038.
Intramural Research Program of the National Institute on Drug Abuse Haass-Koffler, C.L., Akhlaghi, F., Swift, R.M., Leggio, L., 2017. Altering ethanol phar-
(NIDA); and by the National Center for Advancing Translational macokinetics to treat alcohol use disorder: can you teach an old dog new tricks? J.
Psychopharmacol. 31, 812–818.
Sciences (NCATS) grant UH2/UH3-TR000963 (PIs: Drs Lorenzo Leggio Haass-Koffler, C.L., Giovenco, D.E., Lee, M.R., Zywiak, W.H., de la Monte, S.M., Kenna,
and Fatemeh Akhlaghi). The content of this article is solely the re- G.A., Swift, R.M., Leggio, L., 2016. Serum insulin levels are reduced by intravenous
sponsibility of the authors and does not necessarily represent the offi- ghrelin administration but do not correlate with alcohol craving in alcohol-dependent
individuals. Int. J. Neuropsychopharmacol. 19, pyw048.
cial views of the National Institutes of Health. Howard, A.D., Feighner, S.D., Cully, D.F., Arena, J.P., Liberator, P.A., Rosenblum, C.I.,
Hamelin, M., Hreniuk, D.L., Palyha, O.C., Anderson, J., Paress, P.S., Diaz, C., Chou,
M., Liu, K.K., McKee, K.K., Pong, S.S., Chaung, L.Y., Elbrecht, A., Dashkevicz, M.,
Conflicts of interest Heavens, R., Rigby, M., Sirinathsinghji, D.J., Dean, D.C., Melillo, D.G., Patchett, A.A.,
Nargund, R., Griffin, P.R., DeMartino, J.A., Gupta, S.K., Schaeffer, J.M., Smith, R.G.,
Van der Ploeg, L.H., 1996. A receptor in pituitary and hypothalamus that functions in
The authors declare that they have no conflict of interest.

7
M.R. Lee, et al. Neuropharmacology 170 (2020) 107788

growth hormone release. Science 273, 974–977. 2008. Relationship between the hypothalamic-pituitary-thyroid axis and alcohol
Jerlhag, E., 2019. Gut-brain axis and addictive disorders: a review with focus on alcohol craving in alcohol-dependent patients: a longitudinal study. Alcohol Clin. Exp. Res.
and drugs of abuse. Pharmacol. Ther. 196, 1–14. 32, 2047–2053.
Keizer, R.J., Jansen, R.S., Rosing, H., Thijssen, B., Beijnen, J.H., Schellens, J.H., Huitema, Leggio, L., Zywiak, W.H., Fricchione, S.R., Edwards, S.M., de la Monte, S.M., Swift, R.M.,
A.D., 2015. Incorporation of concentration data below the limit of quantification in Kenna, G.A., 2014. Intravenous ghrelin administration increases alcohol craving in
population pharmacokinetic analyses. Pharmacol. Res. Perspect. 3, e00131. alcohol-dependent heavy drinkers: a preliminary investigation. Biol. Psychiatry 76,
Kenna, G.A., Swift, R.M., Hillemacher, T., Leggio, L., 2012. The relationship of appetitive, 734–741.
reproductive and posterior pituitary hormones to alcoholism and craving in humans. Lin, M.T., Chu, P.C., Leu, S.Y., 1983. Effects of TSH, TRH, LH and LHRH on thermo-
Neuropsychol. Rev. 22, 211–228. regulation and food and water intake in the rat. Neuroendocrinology 37, 206–211.
Kluge, M., Riedl, S., Uhr, M., Schmidt, D., Zhang, X., Yassouridis, A., Steiger, A., 2010. Morris, L.S., Voon, V., Leggio, L., 2018. Stress, motivation, and the gut-brain Axis: a focus
Ghrelin affects the hypothalamus-pituitary-thyroid axis in humans by increasing free on the ghrelin system and alcohol use disorder. Alcohol Clin. Exp. Res. 42,
thyroxine and decreasing TSH in plasma. Eur. J. Endocrinol. 162, 1059–1065. 1378–1389.
Kojima, M., Hosoda, H., Date, Y., Nakazato, M., Matsuo, H., Kangawa, K., 1999. Ghrelin is Muller, T.D., Nogueiras, R., Andermann, M.L., Andrews, Z.B., Anker, S.D., Argente, J.,
a growth-hormone-releasing acylated peptide from stomach. Nature 402, 656–660. Batterham, R.L., Benoit, S.C., Bowers, C.Y., Broglio, F., Casanueva, F.F., D'Alessio, D.,
Kong, J., Chuddy, J., Stock, I.A., Loria, P.M., Straub, S.V., Vage, C., Cameron, K.O., Depoortere, I., Geliebter, A., Ghigo, E., Cole, P.A., Cowley, M., Cummings, D.E.,
Bhattacharya, S.K., Lapham, K., McClure, K.F., Zhang, Y., Jackson, V.M., 2016. Dagher, A., Diano, S., Dickson, S.L., Dieguez, C., Granata, R., Grill, H.J., Grove, K.,
Pharmacological characterization of the first in class clinical candidate PF-05190457: Habegger, K.M., Heppner, K., Heiman, M.L., Holsen, L., Holst, B., Inui, A., Jansson,
a selective ghrelin receptor competitive antagonist with inverse agonism that in- J.O., Kirchner, H., Korbonits, M., Laferrere, B., LeRoux, C.W., Lopez, M., Morin, S.,
creases vagal afferent firing and glucose-dependent insulin secretion ex vivo. Br. J. Nakazato, M., Nass, R., Perez-Tilve, D., Pfluger, P.T., Schwartz, T.W., Seeley, R.J.,
Pharmacol. 173, 1452–1464. Sleeman, M., Sun, Y., Sussel, L., Tong, J., Thorner, M.O., van der Lely, A.J., van der
Koopmann, A., Lippmann, K., Schuster, R., Reinhard, I., Bach, P., Weil, G., Rietschel, M., Ploeg, L.H., Zigman, J.M., Kojima, M., Kangawa, K., Smith, R.G., Horvath, T., Tschop,
Witt, S.H., Wiedemann, K., Kiefer, F., 2017. Drinking water to reduce alcohol M.H., 2015. Ghrelin. Mol. Metab. 4, 437–460.
craving? A randomized controlled study on the impact of ghrelin in mediating the Schwartz, T.W., 1983. Pancreatic polypeptide: a hormone under vagal control.
effects of forced water intake in alcohol addiction. Psychoneuroendocrinology 85, Gastroenterology 85, 1411–1425.
56–62. Tamboli, R.A., Antoun, J., Sidani, R.M., Clements, A., Harmata, E.E., Marks-Shulman, P.,
Lee, M.R., Schwandt, M.L., Bollinger, J.W., Dias, A.A., Oot, E.N., Goldman, D., Gaylinn, B.D., Williams, B., Clements, R.H., Albaugh, V.L., Abumrad, N.N., 2017.
Hodgkinson, C.A., Leggio, L., 2015. Effect of functionally significant deiodinase single Metabolic responses to exogenous ghrelin in obesity and early after Roux-en-Y gastric
nucleotide polymorphisms on drinking behavior in alcohol dependence: an ex- bypass in humans. Diabetes Obes. Metab. 19, 1267–1275.
ploratory investigation. Alcohol Clin. Exp. Res. 39, 1665–1670. Tong, J., Prigeon, R.L., Davis, H.W., Bidlingmaier, M., Kahn, S.E., Cummings, D.E.,
Lee, M.R., Tapocik, J.D., Ghareeb, M., Schwandt, M.L., Dias, A.A., Le, A.N., Cobbina, E., Tschop, M.H., D'Alessio, D., 2010. Ghrelin suppresses glucose-stimulated insulin se-
Farinelli, L.A., Bouhlal, S., Farokhnia, M., Heilig, M., Akhlaghi, F., Leggio, L., 2018. cretion and deteriorates glucose tolerance in healthy humans. Diabetes 59,
The novel ghrelin receptor inverse agonist PF-5190457 administered with alcohol: 2145–2151.
preclinical safety experiments and a phase 1b human laboratory study. Mol. Watson, P.E., 1989. Total body water and blood alcohol levels: updating the funda-
Psychiatry (in press). mentals. In: Crow, K., Barr, R. (Eds.), Human Metabolism of Alcohol.
Leggio, L., 2010. Role of the ghrelin system in alcoholism: acting on the growth hormone Pharmcokinetics, Medicolegal Aspects, and General Interest, vol. 1. CRC Press., Boca
secretagogue receptor to treat alcohol-related diseases. Drug News Perspect. 23, Raton, FL, pp. 41–58.
157–166. Zallar, L.J., Farokhnia, M., Tunstall, B.J., Vendruscolo, L.F., Leggio, L., 2017. The role of
Leggio, L., Ferrulli, A., Cardone, S., Malandrino, N., Mirijello, A., D'Angelo, C., Vonghia, the ghrelin system in drug addiction. Int. Rev. Neurobiol. 136, 89–119.
L., Miceli, A., Capristo, E., Kenna, G.A., Gasbarrini, G., Swift, R.M., Addolorato, G.,

You might also like