ArticlePDF Available

Propranolol pharmacokinetics and pharmacodynamics after single doses and at steady-state

Authors:

Abstract and Figures

The duration and extent of cardiac beta-blockade and their relationship to propranolol pharmacokinetics were assessed in nine healthy volunteers. Each subject received 160 mg of regular propranolol (R), 160 mg of sustained-release propranolol (SR) and no drug (control), both as single doses and once daily for 7 days. After single doses and at steady-state, both products caused a decrease in exercise heart rate for at least 24 h, compared to control. The time course of effect was similar to the time course of serum propranolol concentration. The oral clearance of propranolol decreased from single doses to steady-state for both R and SR; however, the difference achieved statistical significance only for R. These changes were reflected in mean accumulation ratios (AUC steady-state 0–24 h/AUC single dose 0-infinity) of 1.49 and 1.68 for R and SR, respectively. The pharmacokinetic data are consistent with a decrease in intrinsic hepatic clearance of propranolol, leading to an increase in bioavailability at steady-state. Despite a two-fold difference in the bioavailability of R and SR, there was no difference in the area under the effect-time curve at steady-state.
Content may be subject to copyright.
Eur J Clin Pharmacol (1987) 32:315-318
European Journal of
Clinical Pharmacology
© Springer-Verlag 1987
Propranolol Pharmaeokinetics and Pharmaeodynamies After Single
Doses and at Steady-State*
R. L. Lalonde, J. A. Pieper, R.J. Straka, M. B. Bottorff, and D. M. Mirvis
Departments of Clinical Pharmacy and Medicine, University of Tennessee, Memphis, Tennessee, USA
Summary. The duration and extent of cardiac beta-
blockade and their relationship to propranolol
pharmacokinetics were assessed in nine healthy vol-
unteers. Each subject received 160 mg of regular
propranolol (R), 160 mg of sustained-release pro-
pranolol (SR) and no drug (control), both as single
doses and once daily for 7 days.
After single doses and at steady-state, both
products caused a decrease in exercise heart rate for
at least 24 h, compared to control. The time course
of effect was similar to the time course of serum
propranolol concentration. The oral clearance of
propranolol decreased from single doses to steady-
state for both R and SR; however, the difference
achieved statistical significance only for R. These
changes were reflected in mean accumulation ratios
(AUC steady-state 0-24 h/AUC single dose 0-infin-
ity) of 1.49 and 1.68 for R and SR, respectively.
The pharmacokinetic data are consistent with a
decrease in intrinsic hepatic clearance of proprano-
lol, leading to an increase in bioavailability at
steady-state. Despite a two-fold difference in the
bioavailability of R and SIL there was no difference
in the area under the effect-time curve at steady-
state.
Key words:
propranolol; pharmacodynamics, phar-
macokinetics, beta-blockade, sustained-release pro-
pranolol
Propranolol has been shown to accumulate upon
chronic dosing to an extent greater than that expect-
ed based on single dose pharmacokinetics (Wood et
* Presented in part at the III World Conference on Clinical
Pharmacology and Therapeutics, Stockholm, Sweden, July 1986
al. 1978). Also, Silber et al. (1983) have demon-
strated a 56% reduction in propranolol's intrinsic
clearance measured at steady-state as the dose was
increased from 40 to 320 mg per day. This change in
intrinsic clearance coupled with metabolite urinary
excretion data, suggests the elimination of propran-
olol is nonlinear over this dosage range. Using these
data, Wagner (1985) has shown that propranolol in-
put rate profoundly affects its bioavailability. The
consequences of propranolol's nonlinear pharmaco-
kinetics and its implications upon cardiac beta-
blockade have yet to be fully investigated. The pur-
pose of this study is to investigate the effects of
acute and chronic once daily oral administration of
regular or sustained-release propranolol and the re-
lationship between the pharmacokinetics, and the
duration and extent of cardiac beta-blockade.
Methods
Nine healthy male volunteers ranging in age from
23 to 39 years (mean age 27 years) and ranging in
weight from 66 to 82 kg (mean weight 75 kg), partic-
ipated in the study. The subjects had no evidence
of previous or current cardiac, respiratory, renal or
hepatic dysfunction. All were nonsmokers and ab-
stained from taking caffeine containing products,
other drugs and alcohol for the duration of the
study. The study was a randomized cross-over com-
parison of three regimens. During Phase I (single
dose phase), each subject received one sustained-re-
lease propranolol (SR) 160 mg capsule (Inderal LA,
Ayerst Laboratories Inc., New York, NY), regular
propranolol (R) 160 mg (two 80 mg Inderal tablets)
and no drug at all (control period).
The doses were administered at approximately
8:00 a.m. and the subjects fasted for 8 h before and
316
4 h after drug administration. At least 7 days passed
before proceeding to the next regimen. After com-
pletion of Phase I, all subjects entered a multiple
dose study (Phase II) using the same randomized
cross-over design. During Phase II, each drug was
taken once daily for 7 days with at least one drug-
free week between each regimen. On the last day of
each regimen during Phase II and on the test days
of Phase I, subjects underwent repeated treadmill
exercise tests. Each test was performed on a motor
driven treadmill at 6 mph, 10% grade for 2.5 min,
immediately before drug administration (time 0 for
control period) and then at 2, 4, 8, 12, 24, 32 and
48 h. Arterial blood pressure and an electrocardio-
gram were recorded immediately after each exercise
and heart rate (HR) was determined over 15 consec-
utive R-R intervals. Percentage reduction in HR was
calculated as the difference between post exercise
control HR and that measured after propranolol in-
gestion divided by the control HR then multiplied
by 100. After the single doses in Phase I and the last
doses in Phase II, blood was collected by direct ven-
ipuncture at 0, I, 2, 4, 6, 8, 12, 24, 32 and 48 h. No
heparin was used in the sample collection. The se-
rum was separated and frozen at -20 °C until anal-
ysis.
Serum concentrations of propranolol and 4-hy-
droxypropranolol were measured by HPLC with
fluorescence detection (Lo et al. 1982). Intra- and
inter-day coefficients of variation for propranolol
and its metabolite ranged from 1.5% to 10.5%, over
the range of concentrations observed in this study,
with a limit of sensitivity of 1 ng/ml. Standard non-
compartmental equations were used to calculate the
area under the serum concentration-time curve
R. L. Lalonde et al.: Propranolol at Steady-State
(AUC), terminal elimination rate constant (k) and
apparent oral clearance (CL). AUC was calculated
to infinity after single doses and over 24 h at steady-
state.
The kinetic parameters were compared by Fried-
man's analysis of variance and the heart rate data
were compared by parametric ANOVA. Post-hoc
tests were done using nonparametric multiple com-
parison and Newman-Keuls tests, respectively
(Conover 1980). Paired comparisons were done us-
ing the Wilcoxon paired-sample test.
Results
The mean serum propranolol concentration-time
curves are shown in Fig. 1 and the individual phar-
macokinetic parameters are summarized in Table 1.
With each product, the AUC over one dosing inter-
val at steady-state was larger than the AUC after a
single dose. The above AUC increase was evident in
8 of 9 subjects with R (p < 0.05). A similar average
AUC increase was evident with SR; however, be-
cause it occurred in only 6 of 9 subjects, the differ-
ence did not achieve statistical significance. The av-
erage AUC increases were 49% and 68% for R and
SR, respectively (p >0.05 between products). The
bioavailability of SR relative to R did not change
significantly between single doses and at steady-
state (0.55+_0.30 vs 0.52+0.11, p >0.05). The k val-
ues were consistently smaller with SR compared to
R (p < 0.05), probably because of continued absorp-
tion with SR. However, there was no significant
change in k values between single dose and steady-
state. After administration of R, the molar ratios of
Table 1. Propranolol pharmacokinetic and pharmacodynamic parameters for the regular (R) and sustained-release (SR) products
Subjects AUC (ng.h.m1-1) k (h -1) CL (ml.min -1 .kg -a) AUC Effect
(% reduction
in HR)
Single dose Steady-state Single dose Steady-state Single dose Steady-state Steady-state
R SR R SR R SR R SR R SR R SR R SR
1 544 294 1516 671 0.205 0.069 0.165 0.063 68.0 125.6 24.4 55.1 479 392
2 990 280 1307 673 0.132 0.107 0.140 0.121 33.9 t19.6 25.6 49.8 467 402
3 317 251 504 245 0.190 0.085 0.178 0.025 102.7 129.7 64.5 132.7 402 192
4 1048 555 1071 454 0.160 0.065 0.149 0.063 30.9 58.3 30.2 71.3 456 356
5 1074 477 1346 1032 0.185 0.058 0.172 0.078 34.5 77.7 27.5 35.9 551 538
6 1296 1455 2204 1065 0.126 0.088 0.149 0.054 26.6 23.7 15.6 32.3 473 437
7 1004 284 1324 574 0.213 0.073 0.219 0.064 35.3 124.8 26.8 61.8 432 581
8 1105 210 1099 547 0.231 0.042 0.181 0.122 36.6 192.4 36.8 73.9 576 422
9 717 563 1026 634 0.152 0.069 0.205 0.092 49.5 63.1 34.6 56.0 265 590
Mean 900 a 486 b 1267 c 655 b 0.177 a 0.073 b 0.173 a 0.076 b 46.5 a 101.7 b 31,8 c 63.2 b 456 a 434 a
+SD (310) (388) (455) (259) (0.037) (0.019) (0.026) (0.032) 24.5) (50.6) (13.7) (32.4) (90) (125)
For each parameter, means with different letters differ at p < 0.05
317
200 -
100
E
~ ~0
Z
o
Z
0
160
"~ 120140 I
~ w..
*R<SR<C
Z .,
R =
SR
< C
tO0
I
,
I
'
' - ' , . ' , ,
I
, , i , , . , , , ,
0 6 12 18 24 30 36 42 48
TIME
(h)
t :
160
E
,<
n-"
120
3=
i
' ' ' " r ' f [ r r l i I [ , t l r , ~ T r r
6 12 t8 24- 30 .36 4.2 48
TIME
(h)
100
r . . , . . , , , , , p , , ~ , , - . i , , 1
0 6 12 18 24 30 36 42 48
TIME (h)
.,,. ***
SR < R < C
** ** SR < R = (3
r I , ' ' " ' ' ' v , r [ i , [ , r i , $ r , [
0 6
12
18 24 50 36 42 48
TIME:
(n)
200
100
~ 30
Z
0
o
Z
0
L3
R. L. Lalonde et al. : Propranolol at Steady-State
Fig. t. Mean propranolol serum concentrations and exercise heart rates (_+ SE.~n = 9) after regular propranolol ([]), sustained-release
propranolol (A) and control (©). The upper panels refer to single dose and the lower panels to steady-state
4-hydroxypropranolol AUC to propranolol AUC
were 0.074+0.032 and 0.037_+0.030 with single
dose and at steady-state, respectively (p < 0.05). Se-
rum concentrations of 4-hydroxypropranolol were
very low or not measurable after administration of
SR.
The heart rates after treadmill exercise are
shown in Fig. 1. During both control periods, the
mean HR were not statistically different between
any time points. After single doses, both proprano-
lol products caused a decrease in HR at each time
point for up to 32 h, compared to control (p < 0.05).
The effect of R was greater than SR at each time
point up to 8 h; then the converse was true at 24
and 32 h. After 48 h, HR were no longer signifi-
cantly different from control. At steady-state, the
time course of HR was quantitatively similar to the
single dose phase. The effects on the heart rate-
systolic pressure product closely paralleled those on
HR. Table 1 shows the area under the effect-time (%
reduction in HR) curve over one dosing interval at
steady-state. Interestingly, there was no statistical
difference in the total effect observed at steady-state
between R and SR.
Discussion
Recently, Wagner (1985) showed how Michaelis-
Menten elimination of propranolol can explain the
more avid first-pass extraction when the drug is ad-
ministered as a sustained-release formulation, there-
by leading to a decrease in bioavailability. We ob-
served an average relative bioavailability of 0.55
after single doses and 0.52 at steady-state, consistent
with the predictions of Wagner. However, it is un-
clear if decreased absorption from the gut may con-
tribute to the lower bioavailability of SR. Both pro-
pranolol products exhibited an increase in AUC
and decrease in CL at steady-state, compared to sin-
gle doses. These changes reflect a decrease in intrin-
sic hepatic clearance of total drug, provided that
propranolol is completely absorbed and totally
eliminated by hepatic metabolism. A decrease in in-
trinsic hepatic clearance would be expected to in-
crease bioavailability at steady-state with little or no
change in the systemic clearance. This is consistent
with the observed increase in peak concentration
(larger than expected based on the half-life and dos-
ing interval) and lack of change in k at steady-state,
compared to single doses. The decrease in AUC ra-
318 R. L. Lalonde et al.: Propranolol at Steady-State
tio of 4-hydroxypropranolol to parent drug further
suggests that at least part of the decrease in intrinsic
clearance is from a reduced clearance to 4-hydroxy-
propranolol. Our data also indicate that at the dos-
age used, R and SR accumulate to a similar degree,
although there was more variability with the SR.
As indicated by the decrease in exercise HR
compared to control, both propranolol formulations
produce cardiac beta-blockade for at least 24 h after
single doses and at steady-state. We were able to de-
scribe the full time course of pharmacologic effects
since the HR had returned to baseline by 48 h. The
time course of effect tended to follow the time
course of propranolol serum concentrations. We al-
so calculated the area under the effect curve, as a
measure of total cardiac beta-blockade over one
dosing interval at steady-state. Despite a two fold
difference in bioavailability between products, the
total effects observed were essentially the same. In
addition, the peak concentration of regular pro-
pranolol increased 25% between single doses and
steady-state without any appreciable change in the
corresponding pharmacodynamic effects. These
discrepancies are probably explained by the nonlin-
ear serum concentration-effect relationship. We
have shown that the Emax model can best describe
the relationship between propranolol concentration
and percentage reduction in exercise HR (Lalonde
et al. 1987). At the higher concentrations achieved
with R, the concentration-effect curve is more flat
leading to smaller increments in effect with increas-
ing concentrations. Therefore, it must be recognized
that differences in cardiac beta-blockade (duration
of action, total effect) between products will be de-
pendent on the dosage regimen, range of concentra-
tions achieved and the particular pharmacodynamic
parameters that describe the concentration-effect re-
lationship in each individual.
References
Conover WF (1980) Practical non-parametric statistics, 2nd edn.
John Wiley and Sons, New York, NY
Lalonde RL, Pieper JA, Straka RJ, Bottorff MB, Mirvis DM
(1987) Pharmacodynamic modeling of propranolol. Clin Phar-
macol Ther 41:156
Lo Man-Wa, Silber B, Riegelman S (1982) An automated HPLC
method for the assay of propranolol and its basic metabolites
in plasma and urine. J Chromatogr Sci 183 : 126-131
Silber BM, Holford NHG, Riegelman S (1983) Dose dependent
elimination of propranolol and its major metabolites in hu-
mans. J Pharm Sci 72:725-732
Wagner JG (1985) Propranolol: Pooled Michaelis-Menten pa-
rameters and the effect of input rate on bioavailability. Clin
Pharmacol Ther 37:481-487
Wood AJJ, Carr K, Vestal RE, Belcher S, Wilkinson GR, Shand
DG (1978) Direct measurement of propranolol bioavailability
during accumulation to steady-state. Br J Clin Pharmacol 6:
345-350
Received: January20, 1987
accepted in revised form: June 10, 1987
Richard L. Lalonde, Pharm. D.
26 South Dunlap Street, Suite 202
Department of Clinical Pharmacy
Memphis, Tennessee 38163, USA
... When administered in multiple doses, these drugs have been found to inhibit their own metabolizing enzymes. That inhibition resulted in a decrease in their hepatic clearance and led to an increase in the AUC at the steady state [63,64]. ...
Article
Full-text available
The law of superposition underpins first-order linear pharmacokinetic relationships. Most drugs, therefore, after a single dose can be described by first-order or linear processes, which can be superposed to understand multiple-dose regimen behavior. However, there are a number of situations where drugs could display behaviors after multiple dosing that leads to capacity-limited or saturation non-linear kinetics and the law of superposition is overruled. This review presents a practical guide to understand the equations and calculations for single and multiple-dosing regimens after intravenous and oral administration. It also provides the pharmaceutical basis for saturation in ADME processes and the consequent changes in the area under the concentration–time curve, which represents drug exposure that can lead to the modulation of efficacy and/or toxic effects. The pharmacokineticist must implicitly understand the principles of superposition, which are a central tenet of drug behavior and disposition during drug development.
Article
Full-text available
Background Observations in people with cerebral cavernous malformations, and in preclinical models of this disorder, suggest that the β-blocker propranolol might reduce the risk of intracerebral haemorrhage. We aimed to evaluate the safety and efficacy of prolonged treatment with propranolol to reduce the incidence of symptomatic intracerebral haemorrhage or focal neurological deficit in people with familial cerebral cavernous malformations. Methods We conducted a randomised, open-label, blinded-endpoint, phase 2 pilot trial (Treat_CCM) at six national reference centres for rare diseases in Italy. People aged 18 years or older with symptomatic familial cerebral cavernous malformation were eligible for enrolment. Participants were randomly assigned (2:1) to receive either oral propranolol (20–320 mg daily) plus standard care (intervention group), or standard care alone (control group), for 24 months. Participants, caregivers, and investigators were aware of treatment group assignment. Participants had clinical assessments and 3 T brain MRI at baseline and at 12 and 24 months. The primary outcome was new occurrence of symptomatic intracerebral haemorrhage or focal neurological deficit attributable to cerebral cavernous malformation over 24 months. Outcome assessors were masked to treatment group assignment. The primary analysis was done in the intention-to-treat population. Because of the pilot study design, we chose a one-sided 80% CI, which could either exclude a clinically meaningful effect or show a signal of efficacy. This trial is registered with EudraCT, 2017-003595-30, and ClinicalTrials.gov, NCT03589014, and is closed to recruitment. Findings Between April 11, 2018, and Dec 5, 2019, 95 people were assessed for eligibility and 83 were enrolled, of whom 57 were assigned to the propranolol plus standard care group and 26 to the standard care alone group. The mean age of participants was 46 years (SD 15); 48 (58%) were female and 35 (42%) were male. The incidence of symptomatic intracerebral haemorrhage or focal neurological deficit was 1·7 (95% CI 1·4–2·0) cases per 100 person-years (two [4%] of 57 participants) in the propranolol plus standard care group and 3·9 (3·1–4·7) per 100 person-years (two [8%] of 26) in the standard care alone group (univariable hazard ratio [HR] 0·43, 80% CI 0·18–0·98). The univariable HR showed a signal of efficacy, according to predefined criteria. The incidence of hospitalisation did not differ between groups (8·2 cases [95% CI 7·5–8·9] per 100 person-years in the propranolol plus standard care group vs 8·2 [95% CI 7·1–9·3] per 100 person-years in the standard care alone group). One participant in the standard care alone group died of sepsis. Three participants in the propranolol plus standard care group discontinued propranolol due to side-effects (two reported hypotension and one reported weakness). Interpretation Propranolol was safe and well tolerated in this population. Propranolol might be beneficial for reducing the incidence of clinical events in people with symptomatic familial cerebral cavernous malformations, although this trial was not designed to be adequately powered to investigate efficacy. A definitive phase 3 trial of propranolol in people with symptomatic familial cerebral cavernous malformations is justified. Funding Italian Medicines Agency, Associazione Italiana per la Ricerca sul Cancro, Swedish Science Council, Knut and Alice Wallenberg Foundation, CARIPLO Foundation, Italian Ministry of Health.
Chapter
The timing of activity of biological molecules has been quantified since the earliest studies on receptors and enzymes. Binding kinetics considers the rate of formation of the drug-target complex (the association rate constant) and the stability of the complex (the residence time, defined by the dissociation rate constant). The last 20 years have seen major advances in our understanding of the significance of binding kinetics to drug activity in in vivo and in vitro, and in the development of new assay technologies to greatly reduce the resource burden of quantifying the time course of drug-target interaction. In this article these advances are reviewed in the practical context of drug discovery goals and processes. Together with online tools, the goal of the review is to enable investigators to assess the value of target binding kinetics to their targets of interest, and to provide a guide for developing and implement kinetic pharmacology assays in drug discovery.
Article
In this review, we evaluate the variability in the pharmacokinetics of 11 drugs with established prophylactic effects in migraine to facilitate ‘personalized medicine’ with these drugs. PubMed was searched for ‘single-dose’ and ‘steady-state’ pharmacokinetic studies of these 11 drugs. The maximum plasma concentration was reported in 248 single-dose and 115 steady-state pharmacokinetic studies, and the area under the plasma concentration-time curve was reported in 299 single-dose studies and 112 steady-state pharmacokinetic studies. For each study, the coefficient of variation was calculated for maximum plasma concentration and area under the plasma concentration-time curve, and we divided the drug variability into two categories; high variability, coefficient of variation >40%, or low or moderate variability, coefficient of variation <40%. Based on the area under the plasma concentration-time curve in steady-state studies, the following drugs have high pharmacokinetic variability: propranolol in 92% (33/36), metoprolol in 85% (33/39), and amitriptyline in 60% (3/5) of studies. The following drugs have low or moderate variability: atenolol in 100% (2/2), valproate in 100% (15/15), topiramate in 88% (7/8), and naproxen and candesartan in 100% (2/2) of studies. For drugs with low or moderate pharmacokinetic variability, treatment can start without initial titration of doses, whereas titration is used to possibly enhance tolerability of topiramate and amitriptyline. The very high pharmacokinetic variability of metoprolol and propranolol can result in very high plasma concentrations in a small minority of patients, and those drugs should therefore be titrated up from a low initial dose, depending mainly on the occurrence of adverse events.
Article
In this second part of the study an optimized propranolol (CAS 318-98-9) extended release matrix tablet based on polyvinylacetate/povidone excipient was tested and compared with an extended release marketed capsule product. Using the USP dissolution testing, the drug release from the matrix tablets was faster in the initial acid stage, but similar in the buffer stage to that of the market capsule product. When evaluated in a pilot bioequivalence study in eight human volunteers, the two products were not bioequivalent according to the FDA bioequivalence criteria. The matrix tablets produced higher Cmax and AUC0-24h than the market capsules. For the developed tablet formulation the higher initial plasma concentrations correlated positively with the faster initial release observed during in vitro testing. Stability study of the matrix tablet formulation indicated a significant reduction in dissolution rates associated with an increase in tablet hardness under accelerated conditions. However, stability testing under ICH long-term conditions did not influence the physical properties and dissolution characteristics of the tablets. Storage of these tablets must be carried out under controlled temperatures not exceeding 25 °C / 60 % RH. Thus, the newly introduced polyvinylacetate/povidone excipient succeeded in developing propranolol extended release matrix tablets.
Article
To evaluate the cardiac and pulmonary effects of siponimod (BAF312) and propranolol co-administration in healthy subjects. Healthy subjects (n = 76) were randomized in a doubleblind manner to receive propranolol at siponimod steady state (group A), siponimod at propranolol steady state (group B), placebo (group C) and propranolol (group D). Pharmacodynamic evaluations included maximum change from baseline in time-matched hourly average heart rate (Emax HR) and mean arterial blood pressure (Emax MABP) over 24 hours postdose, change from baseline in PR intervals, cardiac rhythm, and forced expiratory volume in 1 second (FEV1). Pharmacokinetic and safety parameters were also assessed. Siponimod and propranolol when administered alone resulted in similar HR decrease at steady state. Compared to propranolol alone, the combination at steady state had an additional 6.21 bpm (95%CI: 2.32, 10.11) decrease of mean EmaxHR, a decrease of 5.04 bpm (0.52, 9.56) for group A and 7.39 bpm (2.87, 11.90) for group B. A minor decrease in MABP and a trend towards PR interval increase were noted with co-administration treatment vs. propranolol alone. There were no episodes of second-degree atrioventricular blocks or sinus pauses > 3 seconds. Baseline-corrected FEV1 was reduced by -0.07 L (95% CI: -0.17, 0.03) for group A and -0.05 L (-0.15, 0.05) for group B vs. propranolol alone. There were no cardiovascular adverse events during coadministration treatment. Coadministration of siponimod and propranolol was well tolerated. Bradyarrhythmic effects were less pronounced when propranolol was added to siponimod steady-state therapy compared with siponimod addition to propranolol.
Article
Infantile hemangioma (IH) is a common, benign tumor occurring in up to 10% of white infants. Propranolol has emerged as a front-line therapy for IH. The retrospective study examined the response of propranolol therapy on hemangioma size. Twenty-seven children (4 to 20 weeks old) with IH were enrolled into the study of oral propranolol solution 2 mg/kg per day divided 3 times daily for 10 months. Response was assessed by size measurements at pretreatment and posttreatment. Scoring was stratified into no response, plateau, and regression groups. Secondary outcomes measured were drug compliance and complications. Twenty-seven consecutive patients with IH were treated with propranolol, of whom 67% completed a 6-month therapy. No correlation was demonstrated between tumor size and age at therapy initiation or patient (P = 0.7 and P = 0.7, respectively). A large number of infants responded to therapy (85.2%). Response was first observed sooner in the regression group compared to plateau responders (15.15 ± 8.06 and 20.5 ± 18.42 days, respectively). A significant difference in median pretreatment and posttreatment tumor size was noted (4.50 vs 1.55 cm, P = 0.02). Attrition was secondary to drug-induced side effects, no response, and dosing noncompliance. Propranolol is a safe and effective first-line therapy for problematic IHs. Therapy should show significant response by 2 weeks. If no response is observed by 3 weeks, then other treatment should be sought.
Article
The characteristics of a new Polyvinylacetate/Povidone based excipient, Kollidon® SR were evaluated for application in extended release matrix tablets. The effects of the following formulation and process variables on tablet properties and drug release were tested: Kollidon® SR concentration in the tablet, addition of external binder for wet granulation, presence of an enteric polymer in the matrix, method of manufacturing and compression force. The similarities in release profiles were evaluated by applying the model independent f2 similarity factor. It was found that Kollidon® SR is suitable for pH-independent extended release matrix tablets. A minimum concentration of 30% polymer was necessary to achieve a coherent matrix, able to extend the release of the incorporated drugs. Increasing the Kollidon® SR concentration in the tablet led to a slower drug release. Drug release followed square root of time dependent kinetics, thus indicating a diffusion-controlled release mechanism. The drug release was influenced by the aqueous solubility of the drug. The drug release rate was faster for wet granulation than direct compression, thus making direct compression the method of choice for manufacturing Kollidon® SR extended release systems. It was found that Kollidon® SR was the main release controlling agent in the presence of an external binder or enteric polymer in the matrix. A significant reduction in the dissolution rates associated with an increase in tablet hardness was observed during the stability test under accelerated conditions. The developed propranolol matrix tablets formulation was compared to the reference listed product (Inderal® LA capsules). It was concluded that Kollidon® SR is a potentially useful excipient for the production of pH-independent extended release matrix tablets.
Article
The disposition of propranolol and the formation of its major metabolites, propranolol glucuronide (I), 4-hydroxypropranolol glucuronide (II), and α-naphthoxylactic acid (III), were examined at steady state in four healthy volunteers given oral doses of 40–320 mg/day. The blood to plasma ratio of propranolol was 0.85 ± 0.11 (SD). In all subjects, the average steady-state concentration (Css) of propranolol in plasma increased disproportionately with dose. There was a 1.8- to 2.6-fold difference in the Css between subjects, a 56 ± 20% reduction in the intrinsic clearance, and a 175% increase in the half-life of propranolol over the range of doses administered. The renal clearance was 75.4 ± 17.5 ml/min for I, 130.6 ± 28.3 ml/min for II, and 56.8 ± 13.3 ml/min for III. The formation of I was saturable in three subjects; the Vmax and Km were 103 ± 43 mg/day and 124 ± 46 ng/ml, respectively. In the remaining subject the nonrenal clearance of I was 496 ml/min. The formation of II and III was saturable in all subjects. The Vmax and Km were 71 ± 25 mg/day and 46 ± 22 ng/ml, respectively, for II and 92 ± 35 mg/day and 35 ± 24 ng/ml, respectively, for III. In each subject, the formation clearance associated with the unidentified metabolic pathway(s) (accounting for ∼ 45% of the dose) was best described by a saturable process. The Vmax and Km estimated for this pathway were 212 ± 34 mg/day and 40 ± 12 ng/ml, respectively. These results suggest that the elimination of propranolol is saturable in the human at doses from 40 to 320 mg/day and can be explained only partly by saturability in the metabolic pathways resulting in the formation of I, II, and III.
Article
1. A high performance liquid chromatographic method for the determination of propranolol in human plasma and blood has been developed and used to confirm that cumulation occurred during chronic oral administration, steady-state being achieved within 48 h of beginning 80 mg of the drug every 8 h. 2. The method was adapted to measure [H3]-propranolol and native drug in the same blood sample and was applied to determine simultaneously the disposition of i.v. ([H3]-propranolol) and orally (non-labelled) administered drug after single oral dose of 80 mg and when steady-state had been established on an 80 mg, 8-hourly regimen. 3. Using this approach it was possible to show that a reduced oral clearance at steady-state was associated with a smaller reduction in systemic (i.v.) clearance and no change in liver blood flow. A direct estimate of bioavailability was also possible and was found to be increased at steady-state compared with a single oral dose. 4. We conclude that the accumulation of propranolol during the attainment of steady-state is due to a reduction in intrinsic clearance, resulting in reduced presystemic hepatic extraction.
Article
Average steady-state propranolol plasma concentration (Css) were calculated from published steady-state propranolol clearance data for dose rates (Ro) of 40, 80, 160, 240, and 320 mg/ day in divided doses every 6 hours. The Css-Ro data for each of four subjects were fit essentially perfectly by the equation: Css = KmRo/ (Vm-Ro). Very similar Vm and Km values were obtained with the Vmi and Kmi values for four parallel Michaelis-Menten pathways of propranolol metabolism. It is shown by use of the mean Vm and Km values that the propranolol input rate profoundly affects its bioavailability, which is expected for a first-pass drug that follows Michaelis-Menten elimination kinetics after oral dosing. This most likely explains the poor bioavailability of propranolol after a sustained-release formulation. The decreased bioavailability of propranolol when the number of subdivisions of the daily dose is increased is also explained.
Article
An automated HPLC method is described for the simultaneous determination of propranolol, 4-hydroxypropranolol, and N-desisopropylpropranolol in plasma and urine before and after β-glucuronidase/aryl sulfatase treatment. It involves extraction with ether at pH 10 in the presence of ascorbic acid, added to prevent oxidation of 4-hydroxypropranolol. The compounds are then back extracted into dilute acid and assayed on an HPLC using a fluorescence detector. Three HPLC columns have been used (a phenyl, an octyl, and an octadecyl column). The last column was found to be most reproducible with minimal inter-column variation. The solvent system includes a combination of acetonitrile, methanol, and phosphoric acid. Concentrations as low as 0.2, 1.0, and 0.2 ng/ml of propranolol, 4-hydroxypropranolol, and N-desisopropylpropranolol , respectively, can be measured using 1 ml of plasma.
Pharmacodynamic modeling of propranolol
  • R L Lalonde
  • J A Pieper
  • R J Straka
  • M B Bottorff
  • D M Mirvis
  • RL Lalonde
Lalonde RL, Pieper JA, Straka RJ, Bottorff MB, Mirvis DM (1987) Pharmacodynamic modeling of propranolol. Clin Pharmacol Ther 41:156