Skip to main content
. Author manuscript; available in PMC: 2023 Apr 24.
Published in final edited form as: Prog Neurobiol. 2013 Sep 14;113:40–55. doi: 10.1016/j.pneurobio.2013.08.004

Table 1.

Allopregnanolone (Allo) pre-clinical research translation in AD mouse models and existing clinical exposure data in healthy subjects. Allo blood plasma and brain concentrations and reported safety and efficacy by multiple routes of administration. Intravenous (IV) dose studies determine the pharmacokinetic properties. Assumed to be 100% bioavailable, the IV route of administration is the most quantitative, and useful for gauging alternative routes of administration. Our pre-clinical Allo dosing studies with subcutaneous (SC), transdermal (TD), intranasal (IN), and IV routes of administration that resulted in beneficial effects that decreased hallmarks of AD pathology and improved cognitive performance were tabulated alongside with other pre-clinical studies that tested long-term chronic continuous exposure to SC Allo that resulted in deleterious effects on AD pathology and long-term memory impairment. Human IV dosing is included to compare the mouse pharmacokinetic parameters to clinical Allo pharmacokinetics demonstrates the safety of Allo when blood levels are below the maximum tolerated dose indicated by sedation level. When bioavailability and species-specific allometric conversions are taken into account the pre-clinical studies that demonstrate neuroregenerative efficacy in an AD mouse model are within the safe range of dosage for humans. The AD mouse models were administered both chronic and acute dosing regimens. Allo treatment induced a significant increase in neurogenesis, with the latter regimen yielding the greater increase in neurogenesis. However, the 3/week/3 months (12/month) treatment induced a significant decrease in neurogenesis. Brain sections from 3xTgAD mice treated with Allo or vehicle were immunostained. Aβ immunoreactivity was detected and indicated that the 1/week/6 months (4/month) Allo treatment significantly decreased Aβ immunoreactivity. Efficacy of Aβ reduction in 4/month was comparable to the 12/month Allo treatment whereas the 1/month Allo treatment was ineffective at reducing Aβ immunoreactivity (Chen et al., 2011). Allo tested to mimic chronic stress levels associated with AD. In a series of studies, continuous subcutaneous osmotic pump infusion of Allo proved to be detrimental and decreased learning and memory performance while simultaneously exacerbating AD pathology markers (Bengtsson et al., 2012, 2013). Although not measured, neurogenesis possibly was inhibited in the continuous Allo exposure studies. From these tabulated studies we hypothesized that the dose and frequency of Allo are critical components of efficacy at neurogenic and pathological endpoints.

Purpose Species/Strain Route ALLO mg/kg Vehicle Frequency of exposure Endpoint Blood [ALLO] Brain [ALLO] Results Safety Reference
Pre-clinical Allo pre-clinical efficacy and route of administration development in mouse model of Alzheimer’s disease Mouse (male) 3xTgAD: APPSwe × PS1 M146V × Tau P301L SC 10 PBS/5% EtOH Single dose 24h 4 ng/ml (12.5 nmol/L) C24h 15 ng/g C24h Increased neurogenesis No adverse effects Wang et al. (2010)
SC 10 PBS/5% EtOH Single dose 24 h time course 34ng/ml (107nmol/L) Cmax; 0.5h Tmax 159ng/g C0.5 h Increased neurogenesis No adverse effects Irwin et al. (2013)
IV 1.5 6% HβCD Single dose 24 h time course 215 ng/ml (675 nmol/L) Cmax; 0.08 h Tmax 639 ng/g Cmax Increased CREB signaling and neurodifferentiation markers No adverse effects other than acute mild sedation Irwin et al. (2013)
TD 10–50 Various soluble formulations Single dose 4h; 24h 44 ng/ml (138nmol/L) C4h Increased neurogenesis No adverse effects Irwin et al. (2013)
IN 3–10
SC 10 PBS/5% EtOH Single dose 3 wk Increased neurogenesis and cell survival; improved hippocampal-dependent learning and memory No adverse effects Singh et al. (2011)
SC 10 PBS/5% EtOH Three/wk 3 mo No effect on neurogenesis; decreased β-amyloid No adverse effects Chen et al. (2011)
SC 10 20% HβCD Once/wk 6 mo Increased neurogenesis and cell survival; decreased AD pathology No serious adverse effects; acute hyperactivity followed by brief sedation Chen et al. (2011)
Allo pre-clinical efficacy and regimen development in mouse model of Alzheimer’s disease Mouse APPSwe × PS1ΔE9 (male and female, n = 6–11) SC 1.2–2.4 mg/kg/day osmotic pump 60%SBEβCD Continuous release 3 mo 1.3–2.2 ng/ml (4–7 nmol/L) Cmax wk 4 2.5–4.8 ng/g Cmax wk 4 Chronic treatment for three months increased soluble Aβ1–42; impaired cognitive learning assessed one month after treatment to observe long-term effects Accelerated Alzheimer’s pathology and memory impairment Bengtsson et al. (2012)
Mouse APPSwe × PS1ΔE9 (male and female) SC 2.4–4.8 mg/kg/day osmotic pump 60%SBEβCD Continuous release 1 mo Brief but chronic Allo for one month increased soluble Aβ1–40 and Aβ1–42 but was not sufficient to impair cognitive learning Accelerated Alzheimer’s pathology Bengtsson et al. (2013)
Mouse APPSweArc (male and female) SC 2.4mg/kg/day osmotic pump 60% SBEβCD Continuous release 1 mo Brief but chronic treatment for one month did not affect soluble Aβ1–40 and Aβ1–42 but impaired cognitive learning Memory impairment Bengtsson et al. (2013)
Clinical Clinical trial Human(healthy male, n = 9) Human (healthy female on oral contraceptive, n = 9) IV 0.09 Albumin solution Acute 1 h cumulative doses During 1 h cumulative dosing 48 ng/ml (150nmol/L) Cmax Male – decreased saccadic eye movement; decrease in contentedness No adverse effects other than self reported sedation van Broekhoven et al. (2007)
32 ng/ml (100nmol/L) Cmax Female – decreased saccadic eye movement; increase in contentedness
Clinical trial Human (healthy female, n = 10) IV 0.09 Albumin solution Acute 1 h cumulative doses During 1 h cumulative dosing 22 ng/ml (70nmol/L) Cmax Decreased saccadic eye movement No adverse effects other than fatigue, mild nausea Timby et al. (2006)
Clinical trial Human (healthy female, n = 28 crossover) IV 0.07 Albumin solution Single dose 10–55 min post-dosing 22–45 ng/ml (70–140nmol/L) Cmax Acutely impaired free verbal recall episodic memory due to mild sedation with high variability; no acute effect on semantic or working memory No adverse effects other than self reported sedation Kask et al. (2008)
Clinical trial Human (healthy female, n = 12; PMDD, n = 16) IV 0.05 Albumin solution Single dose 0–20 min post-dosing 16–22 ng/ml (50–70 nmol/L) Cmax No effect on startle response or prepulse inhibition to startle No adverse effects other than self reported sedation Kask et al. (2009)