ArticlePDF Available

Tricyclic antidepressant poisoning: An evidence-based consensus guideline for out-of-hospital management

Authors:

Abstract and Figures

A review of U.S. poison center data for 2004 showed over 12,000 exposures to tricyclic antidepressants (TCAs). A guideline that determines the conditions for emergency department referral and prehospital care could potentially optimize patient outcome, avoid unnecessary emergency department visits, reduce healthcare costs, and reduce life disruption for patients and caregivers. An evidence-based expert consensus process was used to create the guideline. Relevant articles were abstracted by a trained physician researcher. The first draft of the guideline was created by the lead author. The entire panel discussed and refined the guideline before distribution to secondary reviewers for comment. The panel then made changes based on the secondary review comments. The objective of this guideline is to assist poison center personnel in the appropriate prehospital triage and management of patients with suspected ingestions of TCAs by 1) describing the manner in which an ingestion of a TCA might be managed, 2) identifying the key decision elements in managing cases of TCA ingestion, 3) providing clear and practical recommendations that reflect the current state of knowledge, and 4) identifying needs for research. This guideline applies to ingestion of TCAs alone. Co-ingestion of additional substances could require different referral and management recommendations depending on their combined toxicities. This guideline is based on the assessment of current scientific and clinical information. The panel recognizes that specific patient care decisions may be at variance with this guideline and are the prerogative of the patient and the health professionals providing care, considering all the circumstances involved. This guideline does not substitute for clinical judgment. Recommendations are in chronological order of likely clinical use. The grade of recommendation is in parentheses. 1) Patients with suspected self-harm or who are the victims of malicious administration of a TCA should be referred to an emergency department immediately (Grade D). 2) Patients with acute TCA ingestions who are less than 6 years of age and other patients without evidence of self-harm should have further evaluation including standard history taking and determination of the presence of co-ingestants (especially other psychopharmaceutical agents) and underlying exacerbating conditions, such as convulsions or cardiac arrhythmias. Ingestion of a TCA in combination with other drugs might warrant referral to an emergency department. The ingestion of a TCA by a patient with significant underlying cardiovascular or neurological disease should cause referral to an emergency department at a lower dose than for other individuals. Because of the potential severity of TCA poisoning, transportation by EMS, with close monitoring of clinical status and vital signs en route, should be considered (Grade D). 3) Patients who are symptomatic (e.g., weak, drowsy, dizzy, tremulous, palpitations) after a TCA ingestion should be referred to an emergency department (Grade B). 4) Ingestion of either of the following amounts (whichever is lower) would warrant consideration of referral to an emergency department: an amount that exceeds the usual maximum single therapeutic dose or an amount equal to or greater than the lowest reported toxic dose. For all TCAs except desipramine, nortriptyline, trimipramine, and protriptyline, this dose is >5 mg/kg. For despiramine it is >2.5 mg/kg; for nortriptyline it is >2.5 mg/kg; for trimipramine it is >2.5 mg/kg; and for protriptyline it is >1 mg/kg. This recommendation applies to both patients who are naïve to the specific TCA and to patients currently taking cyclic antidepressants who take extra doses, in which case the extra doses should be added to the daily dose taken and then compared to the threshold dose for referral to an emergency department (Grades B/C). 5) Do not induce emesis (Grade D). 6) The risk-to-benefit ratio of prehospital activated charcoal for gastrointestinal decontamination in TCA poisoning is unknown. Prehospital activated charcoal administration, if available, should only be carried out by health professionals and only if no contraindications are present. Do not delay transportation in order to administer activated charcoal (Grades B/D). 7) For unintentional poisonings, asymptomatic patients are unlikely to develop symptoms if the interval between the ingestion and the initial call to a poison center is greater than 6 hours. These patients do not need referral to an emergency department facility (Grade C). 8) Follow-up calls to determine the outcome for a TCA ingestions ideally should be made within 4 hours of the initial call to a poison center and then at appropriate intervals thereafter based on the clinical judgment of the poison center staff (Grade D). 9) An ECG or rhythm strip, if available, should be checked during the prehospital assessment of a TCA overdose patient. A wide-complex arrhythmia with a QRS duration longer than 100 msec is an indicator that the patient should be immediately stabilized, given sodium bicarbonate if there is a protocol for its use, and transported to an emergency department (Grade B). 10) Symptomatic patients with TCA poisoning might require prehospital interventions, such as intravenous fluids, cardiovascular agents, and respiratory support, in accordance with standard ACLS guidelines (Grade D). 11) Administration of sodium bicarbonate might be beneficial for patients with severe or life-threatening TCA toxicity if there is a prehospital protocol for its use (Grades B/D). 12) For TCA-associated convulsions, benzodiazepines are recommended (Grade D). 13) Flumazenil is not recommended for patients with TCA poisoning (Grade D).
Content may be subject to copyright.
Clinical Toxicology (2007) 45, 203–233
Copyright © Informa Healthcare
ISSN: 1556-3650 print / 1556-9519 online
DOI: 10.1080/15563650701226192
LCLT
PRACTICE GUIDELINE
Tricyclic antidepressant poisoning: an evidence-based
consensus guideline for out-of-hospital management*
Tricyclic antid epressant poisoning
ALAN D. WOOLF, M.D., M.P.H., ANDREW R. ERDMAN, M.D., LEWIS S. NELSON, M.D.,
E. MARTIN CARAVATI, M.D., M.P.H., DANIEL J. COBAUGH, P
HARM.D., LISA L. BOOZE, PHARM.D., PAUL M. WAX, M.D.,
ANTHONY S. MANOGUERRA, P
HARM.D., ELIZABETH J. SCHARMAN, PHARM.D., KENT R. OLSON, M.D.,
PETER A. CHYKA, P
HARM.D., GWENN CHRISTIANSON, M.S.N., and WILLIAM G. TROUTMAN, PHARM.D.
American Association of Poison Control Centers, Washington, District of Columbia, USA
A review of U.S. poison center data for 2004 showed over 12,000 exposures to tricyclic antidepressants (TCAs). A guideline that
determines the conditions for emergency department referral and prehospital care could potentially optimize patient outcome, avoid
unnecessary emergency department visits, reduce healthcare costs, and reduce life disruption for patients and caregivers. An evidence-
based expert consensus process was used to create the guideline. Relevant articles were abstracted by a trained physician researcher. The
first draft of the guideline was created by the lead author. The entire panel discussed and refined the guideline before distribution to
secondary reviewers for comment. The panel then made changes based on the secondary review comments. The objective of this guideline
is to assist poison center personnel in the appropriate prehospital triage and management of patients with suspected ingestions of TCAs by
1) describing the manner in which an ingestion of a TCA might be managed, 2) identifying the key decision elements in managing cases of
TCA ingestion, 3) providing clear and practical recommendations that reflect the current state of knowledge, and 4) identifying needs for
research. This guideline applies to ingestion of TCAs alone. Co-ingestion of additional substances could require different referral and
management recommendations depending on their combined toxicities. This guideline is based on the assessment of current scientific and
clinical information. The panel recognizes that specific patient care decisions may be at variance with this guideline and are the prerogative
of the patient and the health professionals providing care, considering all the circumstances involved. This guideline does not substitute for
clinical judgment. Recommendations are in chronological order of likely clinical use. The grade of recommendation is in parentheses. 1)
Patients with suspected self-harm or who are the victims of malicious administration of a TCA should be referred to an emergency
department immediately (Grade D). 2) Patients with acute TCA ingestions who are less than 6 years of age and other patients without
evidence of self-harm should have further evaluation including standard history taking and determination of the presence of co-ingestants
(especially other psychopharmaceutical agents) and underlying exacerbating conditions, such as convulsions or cardiac arrhythmias.
Ingestion of a TCA in combination with other drugs might warrant referral to an emergency department. The ingestion of a TCA by a
patient with significant underlying cardiovascular or neurological disease should cause referral to an emergency department at a lower dose
than for other individuals. Because of the potential severity of TCA poisoning, transportation by EMS, with close monitoring of clinical
status and vital signs en route, should be considered (Grade D). 3) Patients who are symptomatic (e.g., weak, drowsy, dizzy, tremulous,
palpitations) after a TCA ingestion should be referred to an emergency department (Grade B). 4) Ingestion of either of the following
amounts (whichever is lower) would warrant consideration of referral to an emergency department: an amount that exceeds the usual
maximum single therapeutic dose or an amount equal to or greater than the lowest reported toxic dose. For all TCAs except desipramine,
nortriptyline, trimipramine, and protriptyline, this dose is >5 mg/kg. For despiramine it is >2.5 mg/kg; for nortriptyline it is >2.5 mg/kg; for
trimipramine it is >2.5 mg/kg; and for protriptyline it is >1 mg/kg. This recommendation applies to both patients who are naïve to the
specific TCA and to patients currently taking cyclic antidepressants who take extra doses, in which case the extra doses should be added to
the daily dose taken and then compared to the threshold dose for referral to an emergency department (Grades B/C). 5) Do not induce
emesis (Grade D). 6) The risk-to-benefit ratio of prehospital activated charcoal for gastrointestinal decontamination in TCA poisoning is
unknown. Prehospital activated charcoal administration, if available, should only be carried out by health professionals and only if no
contraindications are present. Do not delay transportation in order to administer activated charcoal (Grades B/D). 7) For unintentional
poisonings, asymptomatic patients are unlikely to develop symptoms if the interval between the ingestion and the initial call to a poison
center is greater than 6 hours. These patients do not need referral to an emergency department facility (Grade C). 8) Follow-up calls to
determine the outcome for a TCA ingestions ideally should be made within 4 hours of the initial call to a poison center and then at
Received 20 July 2006; accepted 20 July 2006.
*Guidelines for the Management of Poisoning, supported in full by Cooperative Agreement 8 U4BHS00084 between the American
Association of Poison Control Centers and the Health Resources and Services Administration, Department of Health and Human Services.
Address correspondence to American Association of Poison Control Centers, 3201 New Mexico Avenue NW, Suite 330, Washington,
DC 20016, USA. E-mail: info@aapcc.org
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
204 A.D. Woolf et al.
appropriate intervals thereafter based on the clinical judgment of the poison center staff (Grade D). 9) An ECG or rhythm strip, if available,
should be checked during the prehospital assessment of a TCA overdose patient. A wide-complex arrhythmia with a QRS duration longer
than 100 msec is an indicator that the patient should be immediately stabilized, given sodium bicarbonate if there is a protocol for its use,
and transported to an emergency department (Grade B). 10) Symptomatic patients with TCA poisoning might require prehospital
interventions, such as intravenous fluids, cardiovascular agents, and respiratory support, in accordance with standard ACLS guidelines
(Grade D). 11) Administration of sodium bicarbonate might be beneficial for patients with severe or life-threatening TCA toxicity if there is
a prehospital protocol for its use (Grades B/D). 12) For TCA-associated convulsions, benzodiazepines are recommended (Grade D). 13)
Flumazenil is not recommended for patients with TCA poisoning (Grade D).
Keywords Antidepressive agents, tricyclic/poisoning; Poison control centers/standards; Practice guidelines
Introduction
Statement of the scope of the problem and importance of
the guideline
Tricyclic antidepressant (TCA) poisoning is common in the
United States. In 2004, the Toxic Exposure Surveillance System
(TESS) national database of poison center calls maintained by
the American Association of Poison Control Centers (AAPCC)
recorded 7,430 exposures to amitriptyline, 185 to desipramine,
1,288 to doxepin, 819 to imipramine, 1,152 to nortriptyline, and
26 to protriptyline, as well as 1,152 exposures to “other” or
unknown cyclic antidepressants and 182 exposures to a cyclic
antidepressant formulated with a benzodiazepine or phenothiaz-
ine. Within the 12,234 exposures in these categories, there were
1,351 (9%) exposures in children less than 6 years of age and
3,881 (32%) exposures described as unintentional. A total of
9,324 (76%) exposures were treated in healthcare facilities (1).
The high referral rate reflects the potential toxicity of this class
of pharmaceuticals.
The toxicity of TCAs was apparent relatively soon after
their introduction. One case series reported two deaths in 10
adults hospitalized in 1968 for TCA poisoning (2). A review
of 111 TCA deaths reported to a coroner’s office over 8 years
in the 1970s and early 1980s found that 71% had died in the
field and another 13% were dead on arrival at a hospital (3).
In a 5-year cohort study of 172,598 patients on antidepres-
sants reported in 1995, eight of the 50 patients who commit-
ted suicide did so by antidepressant poisoning (4). Table 1
shows the deaths attributed solely to unintentional exposures
to TCA (no apparent co-ingestants) culled from the TESS
database for 1985–2003. In 2004, there were 85 TCA-related
poisoning deaths reported in the TESS database, all of which
were intentional and/or involved co-ingestants (1). Fifty-one
(60%) of these intentional deaths were associated with
amitriptyline.
TCA poisoning is also a cause of important morbidity. One
poison center-based study reported in 1993 that, over a 2-year
period, TCAs alone accounted for 25% of all overdose-
induced convulsions (5). In a 1995 retrospective study of 388
consecutive patients with TCA poisoning admitted to the
Fernand Widal Hospital in Paris over a 4-year period, 6.2%
developed grand mal seizures, although this figure could
underestimate the risk since some patients had also ingested
benzodiazepines, which have anticonvulsant properties (6).
Appropriate referral to healthcare facilities is critically
important as TCA ingestion can result in convulsions, coma,
life-threatening arrhythmias and cardiac conduction distur-
bances, and death. Despite the frequency and severity of TCA
poisoning, there is little consensus among poison centers on
how patients should be managed in the prehospital setting.
Toxicity of different cyclic antidepressants
A prospective study of 489 patients with TCA overdoses per-
formed in the 1970s (including 203 amitriptyline, 68 imi-
pramine, 27 nortriptyline, 27 trimipramine, 22 clomipramine,
and 21 doxepin cases), found no significant differences in
clinical course between individual antidepressants (7). How-
ever, a cohort study comparing death rates per million pre-
scriptions written in Great Britain showed TCAs such as
amitriptyline and imipramine to be more toxic than other
antidepressants (8). A 6-year review of annual mortality rates
per million prescriptions written for antidepressants in Great
Britain during 1987–92 showed TCAs generally had the
highest mortality rates, with desipramine, amitriptyline, and
imipramine contributing disproportionately to deaths,
whereas there were no protriptyline-related deaths (9).
Another British study of 3,185 fatalities due to antidepres-
sants found doxepin to have a disproportionately higher case
fatality rate (CFR) among the elderly. High mortality rates
were also associated with amitriptyline, trimipramine, imi-
pramine, and clomipramine (10). A 3-year, poison center-
based review of 1,313 patients with TCA poisoning reported
by Wedin et al. in 1986 (11) found that 18% of patients
ingesting imipramine suffered convulsions, which was higher
than other TCAs and similar to convulsion rates found with
overdoses of antidepressants such as amoxapine and mapro-
tiline. A secondary analysis of comparative CFRs of five
TCAs using the TESS database for 1983–2002 found that the
CFR of desipramine was 4–12-fold higher than the CFRs of
amitriptyline, doxepin, imipramine, and nortriptyline (12,13).
Thus, there may be some differences between individual
TCA agents in terms of their potential for producing toxicity.
Pharmacology and pharmacokinetics
The neuropharmacology of TCA action is incompletely
understood. Some of the therapeutic effects of TCAs on
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 205
clinical depression have been attributed to their ability to
deplete norepinephrine (NE) from neuronal presynaptic vesi-
cles, selectively inhibit NE transport, and block NE reuptake.
There are variable inhibitory effects on serotonin uptake-
inactivation as well. TCAs have both central and peripheral
anticholinergic properties. Delayed gastric emptying time
and slowed intestinal peristalsis associated with such effects
could account in part for the variable absorption rates and
delayed clinical toxicity seen in TCA poisoning. TCAs also
affect cardiac conduction by blocking fast inward Na
+
chan-
nels on myocardial cells, analogous to type Ia antiarrhyth-
mic drugs. Such actions can lead to arrhythmias and cardiac
conduction disturbances. Blockade of postsynaptic periph-
eral α-adrenergic receptors decreases preload and vascular
resistance and contributes to the hypotension associated
with TCA use (14).
TCAs have narrow dosage ranges for therapeutic effi-
cacy; higher doses are anticipated to produce adverse
effects. A trial in 173 children and adolescents found that
single doses of 3.75 mg/kg of desipramine produced serum
concentrations of 45 ng/mL and clearance rates of 0.74 L/
kg/hour with no significant age- or gender-related variation
in pharmacokinetics (15). The prolonged clinical effects of
TCAs could be related to slow absorption, their large vol-
umes of distribution (10–50 L/kg), and the enterohepatic
recirculation of both parent compound and metabolites. In
overdose, the half-life for amitriptyline elimination (without
decontamination or enhanced elimination therapies) aver-
ages almost 37 hours and is commonly greater than 60
hours (16).
Therapeutic dosages and uses
Table 2 presents the recommended therapeutic dosage ranges
for the medications covered in this guideline. TCAs are fre-
quently prescribed for depression, panic attacks, severe anxi-
ety, phobic disorders, obsessive-compulsive disorders, eating
Table 1. 1985–2003 TESS TCA deaths (single drug, unintentional)
Age/sex TCA
Dose/serum
concentration Circumstances
13 mo/F I Empty bottle/unk time Coma at home, cardiac arrest in ED, cerebral anoxia. Died 36 hr after admission.
18 mo/M I Whole bottle 30 mins to ED. Seizures, arrest, lavage, lorazepam, dopamine, NaHCO
3
. Hypotension,
widened QRS 5 hr later, seizure, arrhythmias, died 27.5 hr after admission.
30 yr/F I Empty bottle No pulse. DOA.
10 mo/F De 600 mg Coma at home; To ED 90 mins after ingestion. Lavaged, ventricular fibrillation, pH 6.9,
CPR, dead within 1 hr.
11 mo/M De Possibly 1 tablet ED by 1 hr. Ataxic, lavage, activated charcoal, seizure, arrest.
15 mo/M De 500 mg
2556 ng/mL
Coma, hypotension. Intubated, ventilated, dopamine, NE, died 2nd d after admission.
19 mo/F De Unk dose/time
1377 ng/mL
Seizure, cardiac arrest at home; pH 6.9; cerebral edema, ileus, diabetes insipidus, brain dead
4 d after admission. Multiple-dose activated charcoal, NaHCO
3
, epinephrine,
isoproterenol, atropine, Fab, mannitol, furosemide, hyperventilated.
20 mo/M De 800 mg
1600 ng/mL
Seizure, hypotension, widened QRS. Within 2 hr of ED admission, 2 cardiac arrests and died.
Phenytoin, NaHCO
3
, pacer, physostigmine.
20 mo/F De Unk dose Seizure at home, skin grey, intubated, p = 60, DOA.
2 yr/F De 1250 mg
3900 ng/mL
Vomited at home, apnea, cardiac arrest in car, DOA.
2 yr/M De 1250 mg
8000 ng/mL
ED by 45 min. Seizure, bradycardia, arrest. ACLS, intubation/atropine, NE, pacing. Dead at 1 hr.
3 yr/M Do Unk dose/time Coma, cardiac arrest at home. DOA.
5 yr/? Do 900 mg Therapeutic error? Vomited; found dead at home.
84 yr/M Do AOC: 200 mg Coma, respiratory depression.
17 mo/M A Unk dose/time Seizure, pneumothorax. NaHCO
3
, intubated, ventilation, dopamine, NE,
diazepam. Died on 12
th
d.
3 yr/M A 100–125 mg Cyanosis, coma, DOA
5 yr/M A Unk dose/time
A 1150 ng/mL
N 790 ng/mL
DOA. Necrotizing pneumonia & pulmonary edema at autopsy.
5 yr/M N
Unk dose
851 ng/mL
Coma, hyperthermic, hypotensive, aspiration pneumonia, arrhythmias. Intubated, ventilated,
lidocaine, NE, bretylium, NaHCO
3
, dopamine, antibiotics. Multisystem failure over 5 d.
ED: emergency department; Fab: Digoxin Fab anti-digoxin antibodies; CPR: cardiopulmonary resuscitation; AOC: acute on chronic; ACLS: advanced car-
diac life support; DOA: dead on arrival at emergency department; NaHCO
3
: sodium bicarbonate; NE: norepinephrine; Unk: unknown; TCA: I = imi-
p
ramine, De = desipramine, A = amitriptyline, Do = doxepin, N = nortriptyline.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
206 A.D. Woolf et al.
disorders, and attention deficit hyperactivity disorders
(14,17). They have also been used for a variety of other con-
ditions including pain syndromes, insomnia, cataplexy,
migraines, fibromyalgia, chronic fatigue, irritable bowel syn-
drome, and peptic ulcer disease (14,18). Doxepin cream is
used for relief of localized pain and for itching.
Clinical toxicity
Typical side effects seen in patients given therapeutic
doses of TCAs include anticholinergic symptoms, such as
tachycardia, mydriasis, dry mouth, warm flushed dry
skin, delayed gastric emptying, slowed intestinal peristal-
sis or even ileus, urinary retention, and confusion or agi-
tation. Headache, fatigue, anxiety, increased intraocular
pressure, blurred vision, drowsiness, weakness, dizziness,
and restlessness are also common, as are gastrointestinal
complaints including constipation, anorexia, nausea, and
epigastric distress (19). Other adverse effects include
ophthalmoplegia or paralysis of gaze (20,21) and acute
pancreatitis (22).
The cardinal features of severe poisoning include life-
threatening cardiac arrhythmias and conduction disturbances,
Table 2. Current formulations and dosing recommendations for tricyclic antidepressants (309–311)
*†
Amitriptyline HCl tablets (Elavil
, Endep): 10, 25, 50, 75, 100, 150 mg
Adults: 40–100 mg initially and daily maintenance (max 150–300 mg daily)
Adolescents & elderly: 10 mg TID and 20 mg q HS (maximum: 200 mg daily)
Clomipramine HCl capsules (Anafranil): 25, 50, 75 mg
Adults: 25 mg initially to 100 mg maintenance (max 300 mg daily)
Children (10–18 years old): 25 mg initially to 100 mg (or 3mg/kg) maintenance (max 200 mg daily)
Desipramine HCl tablets (Norpramin): 10, 25, 50, 75, 100, 150 mg
Adults: 100–200 mg (max 300 mg daily in hospitalized patients)
Adolescents & elderly: 25–100 mg (max 150 mg daily)
Child (6–12 years old): 1–3 mg/kg/day (max 5 mg/kg/day)
Doxepin HCl capsules (Sinequan): 10, 25, 50, 75, 100, 150 mg; oral concentrate 10 mg (base)/ mL
Adults: 25–75 mg (max 300 mg daily – max single dose: 150 mg)
Child: 1–3 mg/kg/day
Doxepin cream (Prudoxin, Zonalon): 5%; each g = 50 mg doxepin HCl
As directed QID.
Imipramine HCl tablets (Norfranil, Tipramine, Impril, Janimine, Novopramine, Tofranil): 10, 25, 50 mg
Impiramine pamoate capsules (Tofranil-PM): 75, 100, 125, 150 mg
Adults: 75–100 mg daily (max 200 mg daily; 300 mg daily for hospitalized patients only)
Adolescents & elderly: 30–40 mg daily (max 100 mg daily)
Children: >5 yr of age 1.5–2.5 mg/kg/day (max 5 mg/kg/d)
Child (enuresis, in 6–12 years old): 25–50 mg at bedtime
Child (enuresis, in 12 year olds) 25–75 mg (max 2.5 mg/kg)
Nortriptyline HCl capsules (Aventyl, Pamelor): 10, 25, 50, 75 mg; oral concentrate 10 mg/5 mL
Adults: 25 mg TID or QID (max 150 mg daily)
Adolescents & elderly: 30–50 mg daily (max 50 mg daily)
Children 6–12 years old: 1–3 mg/kg/day or 10–20 mg/day
Protriptyline HCl tablets (Vivactil): 5, 10 mg
Adults: 15–40 mg daily in divided doses (max 60 mg daily)
Adolescents & elderly: 15 mg daily initially (max 20 mg daily)
Trimipramine maleate capsules (Surmontil): 25, 50, 100 mg
Adults: 75–100 mg in divided doses (max 150 mg outpatients; 200 mg hospitalized patients)
Adolescents & elderly: 50 mg daily (max 100 mg daily)
Tricyclic Antidepressants in Combination
Perphenazine & amitriptyline HCl tablets (Etrafon, Triavil): 2/10, 2/25, 4/10, 4/25, 4 mg/50 mg
Amitriptyline dosing same as above
Chlordiazepoxide & amitriptyline HCl tablets (Limbitrol): 5/12.5, 10 mg/ 25 mg
Amitriptyline dosing same as above
*Manufacturers specify to use their lower maximum doses in outpatient, unmonitored settings.
Manufacturers do not recommend most tricyclic antidepressants for use in children.
Elavil is no longer marketed in the U.S.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 207
abrupt changes in level of consciousness, coma, convul-
sions, hypotension, and sudden death. Pulmonary complica-
tions (e.g., respiratory depression, sudden apnea, aspiration
pneumonia, adult-type respiratory distress syndrome, and
pulmonary edema) can be life threatening. Other signs of
TCA poisoning include palpitations, tachycardia, hyperten-
sion, ileus, tremors, myoclonus, confusion, delirium, and
lethargy.
Definition of terms used in this guideline
Toxicity from TCAs can occur as a result of a single acute
ingestion, which could be unintentional or intentional, or
with repeated or therapeutic use. An acute exposure might
involve unintentional ingestion of a second therapeutic dose in
a patient already on the drug, unintentional ingestion of some-
one else’s therapeutic dose in a patient not taking a TCA, unin-
tentional ingestion by a child, or intentional ingestion.
This guideline focuses on the ingestion of more than a sin-
gle therapeutic dose. It is known that even therapeutic doses
of TCAs can sometimes cause adverse effects in both adults
and children—some idiosyncratic and some dose-dependent.
However, the focus of this guideline is on the effects of TCAs
in overdose. Articles that were reviewed and found to report
adverse effects related to usual therapeutic doses were
included if they provided useful information on the dose-
response relationship.
For the purpose of this guideline, age groups are defined as
1) children less than 6 years of age and 2) older children and
adults. The older age group is much more likely to attempt
self-harm and to conceal the ingestion. Acute exposures are
defined as those occurring over a period of no more than 8
hours, and chronic exposures are those that occur over a
period of more than 8 hours. The terms “out-of-hospital” or
“prehospital” are defined as the period before a patient
reaches a healthcare facility.
Intended users of this guideline
The intended users of this guideline are personnel in U.S.
poison centers. It has been developed for the conditions prev-
alent in the U.S. While the toxicity of tricyclic antidepres-
sants is not expected to vary in a clinically significant manner
in other nations, the out-of-hospital conditions could be much
different. This guideline should not be extrapolated to other
settings unless it has been determined that the conditions
assumed in this guideline are present.
Exclusions
This guideline does not provide guidance on exposures to some
antidepressants such as maprotiline, amoxapine, and loxapine,
which are heterocyclic compounds with somewhat different
adverse effect profiles. Dothiepin, dibenzipine, melipramine,
prothiaden (dosulepin), and other antidepressants not currently
available in the U.S. are not included in this guideline.
This guideline applies to unintentional ingestions or inges-
tions resulting from medication errors. Poisonings resulting
from intentional abuse or self-harm will all require referral to
an emergency department for evaluation. The likelihood of
self-harm is greatest in adolescent and adult patients, who
might also seek to conceal an overdosed. TCAs have been
implicated in cases of Munchausen syndrome by proxy when
parents purposefully overdose their children chronically to
garner attention from healthcare providers (23,24). Likewise,
TCAs have also been administered with homicidal intent or
in instances of child abuse (25,26). If there is suspicion
concerning the circumstances of poisoning, referral to an
emergency department is of paramount importance.
Objective of the guideline
The objective of this guideline is to assist poison center person-
nel in the appropriate prehospital triage and management of
patients with suspected ingestions of tricyclic antidepressants
by 1) describing the manner in which an ingestion of a tricyclic
antidepressant might be managed, 2) identifying the key deci-
sion elements in managing cases of tricyclic antidepressant
ingestion, 3) providing clear and practical recommendations
that reflect the current state of knowledge, and 4) identifying
needs for research. This guideline applies to ingestion of tricy-
clic antidepressants alone. Co-ingestion of additional sub-
stances could require different referral and management
recommendations depending on their combined toxicities.
This guideline is based on the assessment of current scientific
and clinical information. The panel recognizes that specific
patient care decisions may be at variance with this guideline and
are the prerogative of the patient and the health professionals
providing care, considering all the circumstances involved. This
guideline does not substitute for clinical judgment.
Methodology
The methodology used for the preparation of this guideline was
developed after reviewing the key elements of practice guide-
lines (27,28). An expert consensus panel was established to
develop the guideline (Appendix 1). The American Association
of Poison Control Centers (AAPCC), the American Academy of
Clinical Toxicology (AACT), and the American College of
Medical Toxicology (ACMT) appointed members of their orga-
nizations to serve as panel members. To serve on the expert con-
sensus panel, an individual had to have an exceptional record of
accomplishment in clinical care and scientific research in toxi-
cology, board certification as a clinical or medical toxicologist,
significant U.S. poison center experience, and be an opinion
leader with broad esteem. Two specialists in poison information
were included as full panel members to provide the viewpoint of
the end-users of the guideline.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
208 A.D. Woolf et al.
Search strategy
The National Library of Medicine’s MEDLINE database was
searched (1966 to September 2003) using antidepressive agents,
tricyclic as a MeSH term with the subheadings poisoning (po) or
toxicity (to), limited to humans. MEDLINE and PreMEDLINE
(1966 to September 2003) were searched using amitriptyline,
nortriptyline, imipramine, desipramine, protriptyline, clomi-
pramine, and doxepin as textwords (title, abstract, MeSH term,
CAS registry) plus either poison* or overdose* or toxic*, limited
to humans. This process was repeated in International Pharma-
ceutical Abstracts (1970 to September 2003, excluding meeting
abstracts), Science Citation Index (1977 to September 2003),
Database of Abstracts of Reviews of Effects (accessed Septem-
ber 2003), Cochrane Database of Systematic Reviews (accessed
September 2003), and Cochrane Central Register of Controlled
Trials (accessed September 2003).
The bibliography of the tricyclic antidepressant management
in Poisindex (19) was examined and the abstracts of suitable arti-
cles not previously discovered by the search were reviewed. The
bibliographies of recovered articles were reviewed to identify
previously undiscovered articles. In addition, the chapter bibliog-
raphies in six current major pharmacology and toxicology text-
books (14,29–33) were reviewed for additional articles with
original human data. Published abstracts on TCA overdose pre-
sented at the North American Congress of Clinical Toxicology
between the years 1995–2004 were also reviewed.
Criteria used to identify applicable studies
Published studies that provided original information on the epi-
demiology, pharmacology, toxicology, toxic dose, decision-
making, or management of TCA poisoning were included. Ani-
mal studies were not systematically reviewed for the guideline.
Reviews, letters to the editor, commentaries, and published
information that did not contribute original data were excluded.
Article selection
The recovered citations were entered into an EndNote library
and duplicate entries were eliminated. The abstracts of these
articles were reviewed, looking specifically for those that
dealt with 1) estimations of ingested doses with or without
subsequent signs or symptoms, and 2) management tech-
niques that might be suitable for out-of-hospital use (e.g.,
gastrointestinal decontamination). Articles excluded were
those that did not meet either of the preceding criteria, did not
add new data (e.g., reviews with few references, editorials),
or clearly described only inpatient procedures (e.g., hemodi-
alysis) and forensic analyses without exposure details.
Data extraction
All articles retrieved from the original search were reviewed by
a single, trained, physician abstractor. Each article was
assigned a level-of-evidence score from 1 to 6 using the rating
scheme developed by the Centre for Evidence-based Medicine
at Oxford University (Appendix 2). Single case reports were
classified along with case series as level 4. The complete paper
was then reviewed for original human data regarding the toxic
effects of cyclic antidepressants, or original human data
directly relevant to the out-of-hospital management of patients
with cyclic antidepressant-related toxicity or overdose. Rele-
vant data (e.g., dose, resultant effects, time of onset of effects,
therapeutic interventions or decontamination measures given,
efficacy or results of any interventions, and overall patient
outcome) were compiled into a table and a brief summary
description of each article was written. This full evidence table
is available at http://www.aapcc.org/DiscGuidelines/Guide-
lines%20Tables/TCA%20Evidence%20Table.pdf. The com-
pleted table of all abstracted articles was then forwarded to the
guideline primary author and panel members for review and
consideration in developing the guideline. A list of foreign arti-
cles for which English translations were not available and a list
of articles that could not be located were also forwarded to the
guideline primary author for a decision on whether the article
merited translation and inclusion in the guideline. Every
attempt was made to locate such articles and have their crucial
information extracted, translated, and tabulated. Copies of all
of the articles were made available for reading by the panel
members on a secure AAPCC website. In addition to the com-
plete evidence table of all the abstracted articles, several brief
summary tables were generated to highlight the available data
for various relevant subpopulations (e.g., acute pediatric inges-
tions). These summary tables were also forwarded to the
author and guideline panel members. Finally, a written sum-
mary of the available data was also created and distributed by
the abstractor.
Estimation of doses
In many published case reports of childhood poisonings, only a
total dose of the drug and age of the child are given, without the
child’s weight. In order to compare case reports, a dose per kilo-
gram body weight was estimated by using the child’s age, sex,
and the 95
th
percentile weight using standardized growth charts
(34). If the dose and patient age were given but the patient’s sex
was not reported, the 95
th
percentile for boys at that age was
used. Such calculated doses are shown in italics where appropri-
ate throughout the guideline. Table 4 utilizes this method of dose
calculation to compare case report outcomes.
Guideline writing and review
A guideline draft was prepared by the primary author (listed
first). The draft was submitted to the expert consensus panel
for comment. Using a modified Delphi process, comments
from the expert consensus panel members were collected,
copied into a table of comments, and submitted to the pri-
mary author for response. The primary author responded to
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 209
each comment in the table and, when appropriate, the guide-
line draft was modified to incorporate changes suggested by
the panel. The revised guideline draft was again reviewed by
the panel and, if there was no strong objection by any panelist
to any of the changes made by the primary author, the draft
was prepared for the external review process. External review
of the second draft was conducted by distributing it electroni-
cally to AAPCC, AACT, and ACMT members and the sec-
ondary review panel. The secondary review panel consisted
of representatives from the federal government, public health,
emergency services, pediatrics, pharmacy practice, and con-
sumer organizations (Appendix 3). Comments were submitted
via a discussion thread on the AAPCC web site or privately
through email communication to AAPCC staff. All submitted
comments were stripped of any information that would identify
their sources, copied into a table of comments, and reviewed by
the expert consensus panel and the primary author. The primary
author responded to each comment in the table and his
responses and subsequent changes in the guideline were
reviewed and accepted by the panel. Following a meeting of the
panel, the final revision of the guideline was prepared.
Review of the Evidence
Current poison control center practices
Because the toxic doses overlap with the upper therapeutic
range for many of these drugs, some poison control centers
recommend hospital evaluation after any potentially toxic
TCA exposure. One survey of 44 poison centers (30 respon-
dents or 68%) in 1999 reported that 26 of 30 poison centers
(87%) sent all TCA-poisoned children to healthcare facilities
regardless of dose (the other four poison centers varied in
their thresholds: more than 1.5–5 mg/kg) (35). McFee et al.
(36) reported a second survey of 30 poison centers (22
replies, a 73% return rate) the following year. Of 14 poison
centers setting a threshold dose, six referred all children
ingesting more than 5 mg/kg.
To gather additional evidence for this guideline, a request
was sent out in 2004 to all U.S. poison centers for their cur-
rent TCA poisoning triage guidelines. Table 3 presents a
comparison of current practice of the nine poison centers that
provided guidelines (seven other centers indicated that they
did not have guidelines for TCA management). The range of
doses for hospital referral in acute poisoning was from 3 mg/
kg or “one pill” to as much as the TCA therapeutic dose (5
mg/kg). These results give evidence of the need for consensus
building among poison centers nationally for uniformity in
the approach to the triage of prehospital TCA exposures.
Review of textbooks
Several textbooks noted that TCA ingestions greater than
1000 mg in adults are associated with life-threatening toxicity
Table 3. Comparison of poison center guidance: tricyclic antidepressants, May, 2004
Poison center A B C D E F G H I
Stay-home dose
(child)
5 mg/kg <5 mg/kg one pill <3 mg/kg <5 mg/kg Rx dose (6 yr),
double dose (7–64 yr)
<3.5 mg/kg
Protriptyline <1 mg/kg
Follow-up calls 1–2, 6 hr 1–2, 4–6 hr, prn 2, 6 hr 2, 4, 6 hr 2 hr
AC Yes Yes Yes Yes, remove
pill fragments
Yes Yes Yes Yes Yes
Multi-dose AC No No Yes Yes No Yes
Lavage Yes Yes Yes Yes Yes Yes Yes Yes Yes
Ipecac No No Not preferred No No
NaHCO
3
bolus Yes Yes Yes Yes Yes Yes Yes Yes
NaHCO
3
drip Yes Yes Yes Yes Yes Yes Yes Yes
Target blood pH 7.45–7.50 7.45–7.55 7.45–7.55 7.45–7.55 7.45–7.50 7.45–7.55 <7.55
Symptoms
monitored at home
Minor
drowsiness
No No
Minor
drowsiness
——
Minor drowsiness,
urinary retention
No
Legend: prn = as needed, AC = activated charcoal, NaHCO
3
= sodium bicarbonate, Rx = therapeutic.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
210 A.D. Woolf et al.
(31–33). Some also defined a dose of 10 mg/kg or more as
causing significant toxicity in children (19,29–32). None
made any distinctions between individual TCA drugs in
terms of potentially toxic doses. Several texts specified 6
hours after the ingestion as the period during which the
onset of symptoms is likely (19,30,31,33). Some recom-
mended 6 hours as the observation period for symptoms
(assuming the patient had received oral decontamination)
before a patient with TCA poisoning could be medically
cleared (32,33).
Toxic dose considerations
A precise toxic threshold dose was difficult to determine
from the literature for several reasons: 1) a paucity of good
quality studies (no articles specifically investigated a toxic
dose threshold, and only a small number of articles con-
tained dose-effect information), 2) questions about the
accuracy of the dose estimate, because the TCA was not
prospectively administered and dose information relied on
historical data from a witness or parent, 3) presence of co-
ingestants (e.g., barbiturates, antipsychotics, ethanol) that
could alter the clinical presentation, 4) interindividual dif-
ferences in weight or TCA pharmacokinetics, and 5) a num-
ber of articles involved many patients (some of whom
remaining asymptomatic) in which doses and/or effects
were only reported as ranges, percentages, or means. The
task was made more difficult by the fact that therapeutic
doses are associated with adverse effects (e.g., sedation,
heart rate and ECG interval changes). Despite these limita-
tions, some dose-toxicity information could be gleaned
from the cumulative evidence.
Minimum acute toxic dose in children less
than 6 years of age
In 1967, Steel et al. (37) published a level 4 review of 31
cases of childhood amitriptyline and imipramine poison-
ing and cited a report of a TCA dose at 8 mg/kg as a
threshold associated with severe toxicity. The lowest dose
associated with a fatality in that series was 32 mg/kg,
whereas one child survived despite ingesting a dose esti-
mated at 112 mg/kg. A 1974 level 4 report of 60 children
hospitalized for a one-time TCA poisoning estimated the
minimum lethal dose of amitriptyline or imipramine to be
30 mg/kg, although the authors included the case of a 2½-
year-old child who died within 4 hours of ingesting 15 mg/
kg. While the authors listed 15 mg/kg as the dose associ-
ated with severe symptoms in their case series, they stated
“…children who ingest tricyclics in whatever dosage
should always be admitted…” (38).
Additionally, there was a level 2b article (39), a level 3b
investigation (40), and a number of level 4 or 6 case series
(41–48) in which the exact TCA ingested was not reported
but in which some dose-effect information was available. The
lowest dose of an unidentified TCA associated with any tox-
icity (described as “minor”—probably drowsiness) in a child
was an ingestion of 0.5 mg/kg (47); the lowest specified dose
associated with severe toxicity was up to three times the daily
dose (43); and the lowest quantified dose associated with
severe toxicity was 15–25 mg/kg (44). A level 6 abstract of a
retrospective case series of 48 children less than 6 years of
age with acute TCA poisoning found that only three of 43
children at TCA doses of 5 mg/kg or less developed any tox-
icity at all (described as “minimal”) and that only one of the
remaining five children (doses 5–9.4 mg/kg) became
“sleepy” (45). Two level 4 reports (both retrospective and
with few cases) investigating a toxic threshold for uninten-
tional childhood TCA ingestion found that, while children
ingesting doses of less than 5 mg/kg sometimes developed
mild toxicity such as drowsiness, none developed severe
poisoning (46,47).
From more than 60 published case reports of pediatric
TCA poisoning reviewed (see Table 4), symptoms were
reported at doses as low as 3 mg/kg. The lowest amitriptyline
ingestion associated with mild toxicity was 50 mg and the
lowest dose associated with death was 15 mg/kg (38). The
lowest dose of desipramine associated with severe toxicity
was 100 mg [6 mg/kg] in a 3-year-old (49). The lowest dose
of imipramine associated with coma was up to 75 mg [3 mg/
kg] by a 5-year-old, and the lowest dose associated with con-
vulsions was an ingestion of up to 100 mg [7.5 mg/kg] by a
15-month-old (50). A single case involved the ingestion of
325 mg [16 mg/kg] nortriptyline by a 4-year-old, which
resulted in severe toxicity (48).
Minimum acute toxic dose in patients 6 years
of age and older
Life-threatening symptoms in adults are often seen with TCA
doses in excess of 1000 mg. However, cardiac arrest has been
noted with imipramine doses as low as 200 mg (51). A 22-
year-old woman suffered a myocardial infarction 26 hours
after ingesting 300 mg amitriptyline and 80 mg diazepam.
However, in that case report there was no laboratory verifica-
tion of the exposure and there was no blood or urine screen-
ing to rule out other substance abuse that might have
contributed to a myocardial infarction (52). Bramble et al.
(53) studied 27 acute TCA-poisoned patients prospectively
(level 2b) and found that imipramine doses of 1000 mg were
associated with life-threatening symptoms or death in two
patients, while a patient who ingested 600 mg clomipramine
suffered only mild toxicity. Three patients in that series who
ingested 500–750 mg amitriptyline suffered moderate to
severe symptoms. Of four patients ingesting 500–1200 mg
trimipramine, the lowest dose associated with moderate tox-
icity was 1000 mg. There were several articles in which the
exact TCA ingested was not reported but in which some
dose-effect information was available. There was a single
level 2b article (39), two level 3b investigations (40,51), and
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 211
Table 4. One-time overdoses of tricyclic antidepressants in children less than 6 years of age
Drug
Dose*
(mg/kg) Age sex
Onset of
symptoms
Duration of
toxicity
Arrhythmia
Cardiac arrest
Coma
Hypotension
Pneumonia
Pulmonary
edema
Respiratory
arrest
Respiratory
depression
Seizure
Fatal Ref.
A 81 15 mo F 45 min X X X X Yes 228
A
68 24 mo F X No 42
A 36 23 mo M X X X X X X X No 197
A
31 24 mo F <60 min X No 229
A
19 17 mo F X No 230
A 4 48 mo M 3.5 hr 24 hr N 231
A 42 mo F <180 min 24 hr X No 232
A 36 mo M 240 min X X X No 151
A SR
§
60 mo M No 233
N
16 48 mo No 48
N
30 mo M <120 min X X X X No 232
I 213 24 mo M X X X Yes 163
I 188 30 mo M 60 min Several d X X X X No 173
I 155 30 mo M 10 min
X X X X Yes 234
I 116 35 mo M 30 min 2–3 d X X X X X X No 235
I 100 20 mo M 40 min X X X X No 236
I 90 19 mo M <45 min 48 hr X X X No 237
I 86 36 mo M X X X X X Yes 238
I 70 ? M 105 min X X X X No 42
I 68 30 mo M <14 hr X X X X No 236
I 62 30 mo M 3.5 hr Several d X X X X X No 239
I 61 48 mo F No 240
I 57 24 mo F 30 min X X X X X Yes 241
I 60 30 mo F 5 d X X X X No 232
I 47 36 mo F 2 d X X X X No 242
I 47 30 mo Yes 243
I 44 18 mo F X X X Yes 244
I 40 23 mo M <90 min 2 d X X X X X No 245
I 36 44 mo M 2–3 d X X X X X No 246
I 50 20 mo M <180 min X No 232
I 34 22 mo M <120 min 24 hr X X X No 247
I 32 48 mo M 240 min X X X Yes 150
I 31 30 mo M 30 min Several d X X X X X No 248
I 31 30 mo M 15 min X X X X X X Yes 249
I 30 18 mo F 75 min X X X X No 44
I 26 36 mo F <60 min 4 d X X X X X No 221
I 26 20 mo X X X No 250
I 25 30 mo M 120 min >13 hr X No 251
I 25 18 mo M 90 min X X X X Yes 50
I 25 14 mo F X X X X X X Yes 196
I 25 11 mo M 30 min X X X X X Yes 198
I 23 26 mo F 30 min X X X X X X No 252
I 23 18 mo F 90 min 7 d X X X X X No 253
I 21 21 mo F X No 254
I 20 15 mo F X X X Yes 62
I 15 30 mo 240 min X X X X X X 38
I 12 36 mo F X X No 42
I 8 30 mo X X No 254
I 7.5 15 mo M 120 min X X X No 50
I 5 19 mo M 75 min 24 hr X No 50
(Continued)
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
212 A.D. Woolf et al.
eight level 4 case series (41–44,48,54–56). Among all of these
retrospective investigations, the lowest dose of an unidentified
type of TCA associated with mild toxicity was 200 mg (48), the
lowest semi-quantified dose associated with severe toxicity was
up to three times the daily dose (43), and the lowest quantified
dose associated with severe toxicity was 15–25 mg/kg (44).
Amitriptyline
There were two prospective, randomized, controlled trials
(level 1b) in which a dose of 12.5 mg amitriptyline, given to
healthy adults, was associated with sedation (57,58). In a
level 1b, double-blind study of 12 adults, “auditory vigi-
lance” was impaired within 1½ hours of administration of
6.25 mg amitriptyline and lasted 5 hours (57). There were a
number of articles in which the recognition of the amitrip-
tyline ingestion was retrospective, so there was uncertainty
about the actual amount ingested. Among these were five
level 2b articles (16,53,59–61), a single level 3b article
(62), and five level 4 articles (6,48,63–65). Also, there were
over 50 level 4 or 6 articles in which detailed information
was presented concerning 65 cases (see Table 5). Among
these, the lowest dose of amitriptyline associated with mild
toxicity was 50 mg (38) and the lowest dose associated with
severe toxicity was 300 mg (52).
Clomipramine
There was a single level 2b study (53) and four level 4 case
series or case reports (6,22,48,66) that provided information on
dose-effect relationships for clomipramine. The lowest dose
associated with mild toxicity was 600 mg (53), and the lowest
associated with severe toxicity was 750–1500 mg (22).
Desipramine
There were 10 level 4 articles with individual case informa-
tion on dose-effect relationships for desipramine (54,67–75).
The lowest dose of desipramine associated with severe toxic-
ity was 1000–1200 mg (70).
Doxepin
Some subjects in a self-controlled, level 2b trial of eight adult
volunteers taking doxepin 50 mg after an overnight fast experi-
enced pronounced sedation between 30 minutes and 3 hours
after ingestion (76). There were two level 4 retrospective case
series containing dose-effect information for doxepin (48,65),
and there were eight individual cases reported in seven level 4
articles (71,77–82). Among these, the lowest doxepin dose asso-
ciated with toxicity was in a 23-year-old man who died after tak-
ing nine tablets (tablet strength not reported; total dose could
have been as low as 1.3 mg/kg [90 mg total dose] or as high as
19 mg/kg [1350 mg total dose]) (82). A 24-year-old woman
developed severe toxicity (AV block, hypotension and respira-
tory failure) with eventual recovery after ingesting 425 mg dox-
epin but she had concomitantly taken 2500 mg amitriptyline and
1125 mg desipramine (71). Death was reported after an inges-
tion of up to 1500 mg of doxepin (80). Vohra et al. (65) reported
a 42-year-old woman who became unconscious after ingesting
750 mg doxepin and a 19-year-old who experienced only drows-
iness after ingesting 575 mg doxepin; both survived.
Table 4. (Continued)
Drug
Dose
*
(mg/kg) Age sex
Onset of
symptoms
Duration of
toxicity
Arrhythmia
Cardiac arrest
Coma
Hypotension
Pneumonia
Pulmonary
edema
Respiratory
arrest
Respiratory
depression
Seizure
Fatal Ref.
I
3 60 mo M <60 min 48 hr X No 50
I
24 mo M 6 hr Several d X X No 50
I
24 mo F 180 min 12 hr X X X No 255
I
16 mo F ? X Yes 256
I
15 mo M X X X No 205
De 171 24 mo F <60 min X X X X X Yes 257
De 65 19 mo M 90 min 28 hr X X X X X No 214
De 30 19 mo F X X X Yes 44
De 6 15 mo 49
*Italics indicate estimated dose from the child’s weight at 95
th
percentile for age.
Co-ingestant thioridazine.
A = amitriptyline.
A-SR = amitriptyline, sustained release.
Co-ingestant perphenazine.
N
= nortriptyline.
De = desipramine.
§
Co-ingestant orphenadrine.
I = imipramine.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 213
Table 5.
O
ne-t
i
me over
d
oses o
f
tr
i
cyc
li
c ant
id
epressants
i
n pat
i
ents
6
years o
f
age an
d
o
ld
er
Drug Dose (mg) Age (yr) sex Onset (hr) Duration Survived Ref.
A 10,000 32 F Yes 251
A 9000 46 F 1 Several d Yes 258
A 8000 45 M Yes 219
A
*
6300 30 F No 259
A
*
6000 38 M 7 d Yes 260
A
6000 35 F Yes 204
A 6000 29 M 1 Yes 261
A 5000 30 F 5 d Yes 260
A 3750 23 M 2.5 Yes 262
A
*
3750 25 F 3 3 d Yes 263
A 3000 36 M 2 d Yes 264
A
*
2750 31 F Yes 265
A 2500 42 F 2 36 hr Yes 266
A 2500 70 F 3 Yes 267
A 2200 57 M 16 mo Yes 268
A
*
2000 26 F No 269
A
2000 39 F 4 d Yes 202
A 2000 47 M 2 d Yes 270
A 1875 23 F Yes 86
A 1750 19 F Yes 271
A 1500 30 F Yes 272
A
*
1500 38 M Yes 86
A
*
1300 41 F 1 Several d Yes 273
A
*
1250 – F Yes 265
A
1250 25 M No 274
A
*
1250 56 M No 159
A 1200 42 M Yes 275
A 1050 14 F Several d Yes 276
A
*
1050 17 M Yes 93
A 1000 24 M 2 24 hr Yes 277
A
*
1000 35 F Yes 117
A 1000 61 M 9 Yes 278
A
1000 65 M 2 Several d Yes 279
A 1000 67 M 4.5 48 hr Yes 280
A 950 70 F 4.5 No 280
A 800 53 F Yes 281
A
*
850 67 F 1 3.5 hr No 3
A
*
750 14 M Yes 230
A
750 44 M 16 Several d Yes 155
A 750 45 F 13 d No 259
A 525 8 M Yes 229
A 500 45 F 3 Yes 262
A 350 33 F Yes 117
A
*
300 22 F 36 hr Yes 52
A
*
200 46 F 1 Yes 196
A 150 52 F Yes 21
A
*
13 M Yes 230
A
*
18 M Yes 149
A 22 F Yes 151
A
*
22 F Yes 151
A 24 F Yes 265
A
*
24 F 2 Yes 152
A 25 F Yes 282
A
*
27 F No 182
A 28 F 4 d Yes 260
A 28 F 2 Yes 93
(Continued)
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
214 A.D. Woolf et al.
Table 5. (Continued)
Drug Dose (mg) Age (yr) sex Onset (hr) Duration Survived Ref.
A
31 F 3 d Yes 152
A 33 M No 283
A
*
34 F 7 d No 284
A
*
35 F Yes 149
A
*
38 F 1 No 219
A
*
39 F Several d Yes 190
A
*
39 F Several d Yes 285
A 44 F Yes 286
A 44 M 12 Yes 287
A
*†
44 M 50 hr No 92
A
*
46 F 2 56 hr No 154
A 47 F No 269
A
*
55 F 8 d Yes 288
A
*
56 F 0.25 Yes 222
A 16 F 3 d Yes 226
A 63 M 3 d Yes 213
A/I 1000 9 F 2 wk Yes 289
A/I
*
47 F 2 Yes 222
A/I
*
50 F 6 4 hr Yes 290
A/N
*
34 F 3 40 hr No 2
C
*
15,000 27 M 5 >4 d Yes 66
C 750 48 F 6 2 weeks Yes 22
De 9000 15 F No 73
De 2700 14 F Yes 75
De 2500 40 F Yes 54
De 2000 24 F 4 Yes 69
De 1800 22M 7 19 hr Yes 72
De
*
1500 38 F 2 No 68
De 1500 19 F 4 Yes 67
De
*
1200 18 F 12 Yes 70
De 1150 58 F 1.5 Several hr Yes 74
De 19 F Yes 189
De 31M Yes 218
De
*
–27 F No2
Do
*
3000 55 M Yes 77
Do
*
2500 24 M 1 No 78
Do 1500 35 F 3 No 80
Do 1500 54 F 1 24 hr Yes 81
Do
*
600 34 M 4 Yes 79
Do 18 F 2 No 152
Do 23 M 1 13 hr No 82
Do
*
49 F Yes 291
Do
*
53 F Yes 117
I
*
10,000 34 F No 54
I
*
5375 21 F 3.5 Several d Yes 292
I 5350 23 F 1.5 Yes 171
I 4700 41M Several d Yes 293
I 4500 25 F Yes 188
I 2500 30 F Yes 294
I 2500 14 F 2 Yes 211
I 2250 19 F 0.66 Several d Yes 295
I 1875 38 F No 296
I
*
1500 14 F Yes 199
I 1500 29 F 8 3 d Yes 297
I
1250
19 F 0.75 Yes 209
(Continued)
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 215
Imipramine
There was one level 1b, prospective, randomized trial in which a
single 12.5 mg dose of imipramine given to healthy adults
resulted in sedation and dry mouth (83). There were a number of
articles in which the recognition of imipramine ingestion was
retrospective. There was a single level 2b article (53), a single
level 3b article (51), two level 4 case series (38,84), and 30 cases
in 28 level 4 or 6 case reports, case series, or their abstracts. The
lowest dose of imipramine associated with toxicity was 100 mg
(38); the lowest fatal dose was 200 mg (51).
Table 5. (Continued)
Drug Dose (mg) Age (yr) sex Onset (hr) Duration Survived Ref.
I 1250 24 F Yes 136
I
*
1150 38 F 2 Yes 298
I
1000 6 M 4 Yes 299
I
1000 8.5 M No 300
I
*
1000 47 F No 159
I
800 32 M Yes 133
I 750 7 M 4 Yes 300
I 750 36 F No 301
I
625 28 F 2 No 302
I 475 7 F Yes 303
I 10 M No 210
I 18 M No 54
I 24 F No 304
I 25 F Yes 305
I 27 F 1.5 Yes 306
I 27 F Yes 212
I 28 M 1 No 209
I 29 F Yes 54
I 34 F 10 Yes 307
I 36 F No 137
I 37 F No 137
I
*
40 M Yes 308
I 49 F 5 d No 153
I 54 F Yes 200
N
8000 29 F 0.25 Yes 90
N
5000 52 F Yes 89
N
2350 69 F 2 10 hr No 82
N
2000 30 F 17 hr No 91
N
*†
1250 21 F No 92
N
1250 59 M Yes 86
N
950 19 F 0.75 Yes 87
N
*
600 25 F 4 24 hr Yes 85
N
–16 F No88
N
34 F No 151
P 28 M No 201
T
*
3500 21 F Yes 93
T
*
–59 F Yes93
Unk
*
39 F Yes 207
Unk 45 M Yes 208
*Co-ingestant confirmed.
Acute-on-chronic poisoning.
A = amitriptyline.
C = clomipramine.
De = desipramine.
Do = doxepin.
I = imipramine.
N
= nortriptyline.
P = protriptyline.
T = trimipramine.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
216 A.D. Woolf et al.
Nortriptyline
There was a single level 1b, prospective, randomized trial in
which single nortriptyline doses of 12.5 mg given to healthy
adults were associated with sedation, impairment in reaction,
and poor performance of complex tasks (57). There were
several articles in which the recognition of nortriptyline inges-
tion was retrospective. There was a single level 2b article (85),
two level 4 case series (63,84), and nine individually reported
cases (82,85–92). Among these, the lowest dose of nortrip-
tyline associated with severe toxicity was 600 mg (85).
Trimipramine
There was a single level 2b study (53), a single level 4 case
series (6), and a single case report (93) of trimipramine
ingestion. Among these, the lowest dose of trimipramine
associated with moderate toxicity was 1000 mg (53) and the
lowest dose resulting in severe toxicity was 1200 mg (6).
Protriptyline
There are few studies of protriptyline toxicity. In one level 1b
adult study, some functions (tapping rate, arithmetic function,
and reaction time) were perturbed with 25 mg doses of ami-
triptyline and nortriptyline, whereas a therapeutic dose of
protriptyline (10 mg) produced no functional disturbances.
Both nortriptyline and protriptyline (secondary amines) were
associated with much less sedation than amitriptyline (ter-
tiary amine) at such doses (57).
Chronic therapeutic TCA dosing in children less
than 6 years of age
Therapeutic doses of TCAs produce severe symptoms of toxicity
in some children. One child in a prospective trial (level 1b) of imi-
pramine developed listlessness and constipation on 50 mg/day
(94). In a level 3b study, children treated with desipramine (mean
dosage 3 mg/kg/day) had increases in the PR intervals on their
ECGs (95). A 3-year-old boy with autism was treated with
amphetamines and imipramine (25 mg three times daily, increased
to 50 mg three times daily after several weeks). One week after the
upward dose revision, he developed tremors, convulsions, and
then hypotonia, which abated after the medications were discon-
tinued (96). Another 3-year-old with autism also developed con-
vulsions 2 days after reinitiating imipramine treatment at 75 mg
daily. He had previously been on the drug at 125 mg daily for an
unspecified period but had had a 15-day hiatus. He continued to
have convulsions throughout his early life, despite discontinuation
of the imipramine and initiation of anticonvulsant treatment (97).
Chronic therapeutic TCA dosing in patients 6 years
of age and older
Therapeutic dosages of TCAs have adverse effects on con-
sciousness, cognitive function, and cardiovascular status. There
was a single level 1b prospective trial in which mean dosages of
3 mg/kg/day of either clomipramine or desipramine were associ-
ated with mean increases in heart rate and prolonged PR, QRS,
and QTc intervals (98). In a prospective, unblinded trial of imi-
pramine’s effects on depression in 44 adults, patients receiving
3.5 mg/kg/day in divided doses (mean 245 mg daily in men and
218 mg daily in women) experienced orthostatic hypotension,
dizziness, ataxia, and falls (99). A meta-analysis by Wilens et al.
(100) (level 1a) aggregated 24 studies involving 730 children
and adolescents given imipramine, desipramine, or amitriptyline
at doses of 0.7–5 mg/kg (mean 3.7 mg/kg) or nortriptyline at
doses of 0.6–2 mg/kg and found only minor effects on ECG con-
duction parameters, heart rate, and blood pressure. However,
therapeutic doses of TCA taken chronically have been associ-
ated with cardiovascular complications and sudden death in
older children and adolescents (100–106).
Amitriptyline
Two level 1b trials (107,108) and several level 2b prospective
trials (65,109–112) examined amitriptyline given prospectively.
Among them, the lowest dosage associated with toxicity was
100–200 mg/day, resulting in increased heart rate and the pro-
longation of ECG intervals (110). There were also eight reports
of chronic amitriptyline toxicity in five level 4 articles (110,114–
117). Among these, the lowest dosage associated with toxicity
was 50 mg/day, resulting in pedal edema and adynamic ileus
(110). A 73-year-old woman was erroneously prescribed 100
mg tablets instead of 10 mg of amitriptyline (200 mg daily
instead of 20 mg daily) and developed hallucinations, dysarthria,
and incoordination, reversed by physostigmine after several days
(117). Therapeutic dosages have been associated with deaths in
adults. A 60-year-old woman was started on amitriptyline for
depression at a dosage of 25 mg three times daily for 2 weeks (in
addition to phenobarbital, chlorpromazine, and other drugs).
When the dosage was increased to 50 mg three times daily, she
developed abdominal distention, paralytic ileus, and cardiovas-
cular collapse and died (113).
Clomipramine
There were two level 2b trials in which clomipramine was
given prospectively and in which toxic effects developed
(118,119). The lowest dosage of clomipramine associated
with toxicity in adults was 150 mg/day (119). In a level 2b
open-label trial of clomipramine given for the treatment of
autism with movement disorders, three of five children aged
7–12 years on dosages of 3.9–9.8 mg/kg/day developed agita-
tion and aggression requiring hospitalization. Symptoms
abated upon discontinuation of the drug. The authors recom-
mended that children should not receive doses of clomi-
pramine greater than 3–5 mg/kg/day (118).
Desipramine
There were several articles identified in which
desipramine was given prospectively, resulting in toxic
effects. In a level 1b prospective trial, dosages of 100–200
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 217
mg/day were associated with a mean increase in heart rate
and various ECG intervals (108). Rapoport et al. (120)
described a prospective study of 20 boys with enuresis
given 75 mg desipramine nightly for 24 days who reported
higher rates of dry mouth and daytime drowsiness than
placebo-treated controls. Schroeder et al. (121) studied 20
children taking desipramine titrated to a maximum daily
dose of 5 mg/kg (average dose 4.25 mg/kg) for eating dis-
orders, ADD, or affective disorders and found an 18%
increase in heart rate and 9% increase in QTc interval on
ECG at 8 weeks into the study, neither of which was clini-
cally significant. However, seven of 20 children did not
reach the maximum dosage.
There were also two level 4 retrospective case series
(122,123) and several case reports. Wagner and Fershtman
(124) described a 12-year-old boy diagnosed with depression
taking desipramine (2.5 mg/kg/day) who developed symp-
tomatic QTc prolongation (9.7% increase over his baseline)
during 2 months of therapy. Varley and McClellan (105)
reported the sudden deaths of two children—a 9-year-old 5
weeks after starting desipramine (3.3 mg/kg/day) for depres-
sion and a 7-year-old taking a combination of imipramine
(6 mg/kg/day) and thioridazine (1 mg/kg/day) chronically
for a conduct disorder. A 2001 report by Varley (125) identi-
fied eight cases of sudden death during TCA use in children,
including six deaths related to desipramine. A level 4 case
series of three deaths involved children 8 (two cases) and 9
years of age taking desipramine (50 mg daily for one and
unknown dosages for the other two) for periods ranging from
4–6 weeks to 2 years as therapy for attention deficit hyperac-
tivity disorder (ADHD, two cases) or depression. Serum des-
piramine concentrations were only 10 μg/L (two cases) and
85 μg/L (123). A fourth case reported later was a 12-year-old
girl with ADHD. Her dosage of desipramine was raised from
125 mg daily taken during the preceding 6 months to 150 mg
daily in divided doses; she was found unconscious a few days
after the dosage change and died (126).
Doxepin
There were two level 2b prospective trials in which toxic
effects developed when doxepin was given (111,112).
Between them, dosages as low as 150 mg/day were associ-
ated with a higher mean heart rate, and dosages of 200 mg/
day were associated with ST-T changes on ECG. There was
also a level 4 case series in which one patient developed
convulsions while taking 250 mg/day of doxepin (122).
Imipramine
There were nine articles in which toxicity developed when imi-
pramine was given prospectively. Three of these were level 1b
(120,127,128) and six were level 2b (99,109,112,129–131).
Among these prospective trials, the lowest dosages of imi-
pramine associated with toxicity were 50–200 mg/day, which
were associated with agitation, flushes, and insomnia (129). In
one prospective study of 22 children aged 5–17 years, imi-
pramine titrated to a maximum dosage of 5 mg/kg/day in
divided doses produced increases in the QRS complex duration
on ECG in 19 children and a 10% drop in standing systolic
blood pressure in three (130). Rapoport et al. (120), in a pro-
spective, placebo-controlled study of 20 boys ages 7–12 years
old with enuresis treated with 75 mg imipramine nightly for 24
days, reported increased rates of daytime drowsiness, head-
ache, and dry mouth with imipramine.
There was also one level 3b study (95) and two level 4 retro-
spective case series (122,132) along with 16 individual cases of
chronic imipramine toxicity in nine level 4 articles
(96,105,110,133–138). Bartels et al. (95) reviewed the ECGs of
39 children and adolescents before and after starting imipramine
or desipramine therapy (mean maintenance dosages of imi-
pramine and desipramine were 3 and 2.9 mg/kg/day, respec-
tively) and found increases in the PR intervals of 11 patients and
new first-degree AV block in two patients. In another report, two
children, aged 6 and 8 years with hyperactivity, developed new-
onset convulsions after taking imipramine (75 mg three times
daily) for weeks to months (96). The lowest dosage associated
with toxicity in an adult was 75 mg/day, resulting in syncope,
bradycardia, and asystole after 5 days of therapy in a 37-year-old
man. However, he was on other medications and had a history of
glomerulonephritis, hypertension (treated with guanethidine and
methyldopa), and congestive heart failure (138).
Nortriptyline
There were five articles in which nortriptyline was given
chronically and in which toxic effects developed. Four of
these were level 2b (65,112,139,140) and one was a letter
to the editor describing an unpublished study in which
dosages of 50–150 mg/day were associated with cardiac
conduction defects (141). Two level 4 case reports
described chronic nortriptyline toxicity after 2–5 doses of
200 mg/day (142).
Trimipramine
There were two articles in which toxic effects developed
when trimipramine was given chronically. One of these was a
level 1b quality randomized trial (94) and the other was a
level 2b trial (109). Dosages of 50 mg/day were associated
with minor adverse effects including nausea, vomiting,
drowsiness, and rash (94).
ECG diagnosis
Analyses of ECG changes early in TCA poisoning have
been undertaken in order to attempt to predict the risk of
life-threatening cardiovascular or neurological complications.
In a 5-year retrospective study (level 2b) of 225 TCA over-
doses admitted to an intensive care unit, Hulten and Heath
(143) found that patients with QRS durations longer than 100
msec were more likely to develop respiratory depression,
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
218 A.D. Woolf et al.
convulsions, and death. Boehnert and Lovejoy (144) carried
out a level 2b prospective observation study of 49 patients
with acute TCA poisoning presenting within 16 hours of their
overdoses. The 36 patients whose QRS durations exceeded
100 msec had a higher risk of developing convulsions or
arrhythmias. Niemann et al. (145) studied 11 patients with
confirmed TCA poisoning (level 2b) and found that the ter-
minal 40 msec vector on the ECG was the best predictor of
toxicity, although both QRS duration and QTc duration were
also abnormal. Wolfe et al. (146) reviewed (level 2b) the
records of patients with TCA (N = 48) versus other poisoning
(N = 30) and found that a terminal 40 msec axis of 120–270
degrees on the ECG had the highest sensitivity (83%) and
specificity (63%) in predicting TCA toxicity. Liebelt et al.
(147), in a prospective cohort study (level 2b) of 79 TCA
overdoses reported to one poison center, found that an R-
wave greater than 3 mm in lead aVR of the ECG was predic-
tive of both convulsions and dysrhythmias with better sensi-
tivity (81%) and specificity (73%) than either the serum TCA
concentration or QRS duration. A recent systematic review
(level 2a) included 18 studies of the predictors of clinical
severity (arrhythmias, convulsions, or death) in TCA poison-
ing. QRS duration and blood concentration had only fair sen-
sitivity (0.69 and 0.75) and specificity (0.69 and 0.72) in
predicting convulsions and similar sensitivity (0.79 and 0.78)
and worse specificity (0.46 and 0.57) in predicting arrhyth-
mias (148). The panel determined that ECG results are avail-
able from EMS personnel to some poison centers and chose
to consider this parameter for the recommendations offered in
this guideline.
Onset of effects and duration of monitoring
Panel members expressed interest in taking into account the
time for toxicity to develop after TCA exposures, in order to
help make decisions about prehospital transportation and man-
agement. All articles were searched for evidence documenting
or estimating a time of onset. Unfortunately, the vast majority
reported times of presentation to healthcare facilities but not
times of symptom onset, which might have occurred earlier.
Thus, in most cases, it was only possible to establish an upper
limit of time to onset. Care had to be taken when evaluating
these articles not to confuse onset of any adverse effects with
onset of serious or major effects, time to peak effects, or the
occurrence of delayed effects or deterioration. Decontamina-
tion measures might have affected times of onset of symptoms.
It was often not clear whether, or what, symptoms were present
before the late-occurring events emphasized in the reports.
There were few data to distinguish time of onset by patient age
or by individual TCA.
Delayed clinical deterioration
Many patients who developed clinical effects of TCA poison-
ing went on to develop more serious effects later. Those who
did experience significant deterioration tended to do so quite
rapidly, indeed catastrophically in some cases. For example,
Ellison et al. (54) reviewed the charts of 30 patients who
experienced convulsions after TCA overdose (level 4). Con-
vulsions started within 1½ hours of admission in 28 of 30,
23% had been fully alert just prior to the convulsion, and
10% died. Patients in other reports deteriorated progressively
over the course of hours to days after the ingestion
(50,66,68,85,92,149–152). There were also several level 4
reports of delayed toxicity (especially conduction defects or
dysrhythmias) or even death after a period of relative
improvement or stabilization (2,52,66,92,153–155). It is
unclear whether the delayed effects represented recurrence of
direct TCA toxicity (e.g., due to ongoing absorption, inade-
quate decontamination, mobilization of tissue stores) or sec-
ondary complications of hospitalization or treatments (e.g.,
aspiration of gastric contents, adult-type respiratory distress
syndrome), other co-ingestants or co-medications (e.g., ace-
taminophen toxicity, alcohol withdrawal), or a different
pathophysiologic process (e.g., serotonin syndrome, intesti-
nal obstruction, neuropathy due to TCA effects). In some
cases, this deterioration occurred days after the ingestion;
however, in all cases, significant initial toxicity had been pre-
viously documented.
Time of onset
In most cases, the time of effect onset, when noted, appeared
to be within 2 hours after the TCA ingestion. However, in
some, effects did not appear for several hours. Effects were
reported up to 5 hours after amitriptyline or nortriptyline
ingestion in one level 1b therapeutic trial (57).
A 7-year, level 2b review of 88 TCA poisoned patients
found that none developed hemodynamic complications after
12 hours following ingestion (56). A level 4 retrospective
case series of 60 childhood poisonings involving amitrip-
tyline and imipramine reported that symptoms usually devel-
oped within 4 hours of ingestion (38). The longest
documented time to onset of toxicity was 6 hours after an
ingestion of imipramine by a young child (50). In one report
(level 4), a 4-year-old boy with enuresis drank 90 mL imi-
pramine syrup (32 mg/kg), was given a salt-water mixture by
his mother, vomited, “went to sleep,” and 4 hours later devel-
oped convulsions and then ventricular tachycardia and fibril-
lation (150). A 44-year-old man taking 75 mg amitriptyline
daily for months to treat depression ingested 750 mg and pre-
sented to an emergency department 16 hours later with only
drowsiness as a symptom of toxicity (155).
A description of 111 TCA overdose-related deaths (level
4) reported to a coroner’s office reported that most patients
developed major signs within 3 hours of hospital presenta-
tion. Two patients had cardiac arrests more than 3 hours after
presentation; however, the presenting signs and at what time
they arrested were not detailed in the article (3). In a level 2b
review of 102 adults admitted with TCA overdose, the first
manifestation of convulsions and ventricular dysrhythmias
occurred within 6 hours of admission in all patients (156). In
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 219
another level 2b prospective observational series of 49
patients, all convulsions and dysrhythmias occurred within 6
hours of ingestion (144). In a level 2b prospective observa-
tional series of nine adults with severe amitriptyline poison-
ing, the onset of all convulsions, dysrhythmias, and
hypotension occurred within 4 hours of admission (157).
Duration of symptoms
In a level 4 prospective case series of 24 adults admitted with
TCA overdose, two patients developed transient supraven-
tricular tachycardia 2–4 days after ingestion (92). In a retro-
spective review of 316 patients admitted with TCA overdose
(level 4), hypotension and convulsions more than 24 hours
after ingestion were unusual but occurred in two patients and
one patient, respectively (158). In a level 3b observational
series of 40 patients with TCA overdoses, all major effects
occurred within 24 hours of ingestion (40). Similarly, in a
level 2b study of 72 patients admitted with TCA overdose, all
clinical effects, with the exception of aspiration symptoms,
developed within 24 hours of ingestion (83). In a level 4 case
series of 62 adults admitted with TCA overdose, none devel-
oped a new dysrhythmia after 24 hours (159). In a level 3b
report of 40 patients admitted with TCA overdose, maximal
clinical effects and TCA concentrations developed within 24
hours in all patients except one who was on a corticosteroid
for a neurological disorder at the time (160). In a description
(level 4) of 45 patients hospitalized for TCA overdose (60%
of whom presented within 6 hours of the ingestion) no com-
plications occurred after 24 hours. Of note, one patient pre-
sented without any major signs or symptoms but within 6
hours of presentation was comatose (161).
A report (level 4) of 24 adults with convulsions after TCA
overdose found that the onset of convulsions occurred within
18 hours in all patients (6). In a level 4 retrospective review
of 30 patients admitted with convulsions after TCA overdose,
the onset of convulsions ranged from 0.5 to 9.3 hours. Many
of these patients had decreased levels of consciousness prior
to convulsion onset but 23% were awake at the time. Whether
other effects were present in these patients—clinically or
electrocardiographically—is not clear (54). In a level 2b
review of 72 adults admitted to an ICU with TCA overdose,
only one patient developed a new ECG abnormality after
admission (162). A larger level 4 retrospective review of 295
patients admitted to one ICU with TCA overdose found that
multiple patients developed convulsions, circulatory impair-
ment, or ECG abnormalities after admission (48). In a 3-year,
level 2b retrospective review of 75 hospitalized patients with
TCA overdoses, Goldberg et al. (83) found that none of those
who had not aspirated developed any serious new toxicity
more than 24 hours after ingestion. In a level 4 retrospective
review of 38 patients hospitalized for TCA overdoses, all
ECG changes normalized within 24 hours. The authors sug-
gested that patients fully recovered from TCA-related ECG
changes need be monitored only for 12 hours (163).
Emerman et al. (164) performed a 10-year, hospital-based,
level 2b review of the records of 92 adults with TCA over-
dose and found that their initial level of consciousness (Glas-
cow Coma Scale <8: 86% sensitivity, 89% specificity) best
predicted the 37 patients (40%) who developed serious com-
plications—73% developed complications within 30 minutes
of presentation to the emergency department and none devel-
oped new complications more than 2 hours after arrival at the
hospital. In a prospective study of 67 patients, Foulke (165)
concluded that high-risk features predicting late complica-
tions included QRS duration, the presence of arrhythmias or
conduction delays on ECG, altered mental status, convul-
sions, respiratory depression, or hypotension.
Duration of monitoring of asymptomatic patients
The duration of monitoring recommended varies for treated
patients with TCA poisoning. Foulke et al. (156) reviewed
165 TCA overdoses (level 2b) and found that patients with-
out major evidence of toxicity in the emergency department
did not develop serious complications later. Pentel and Sioris
(166), in a level 4 review of 129 adults with TCA overdoses,
found that all who developed neurological or cardiovascular
complications did so within 1 hour of their admission. In their
survey of 30 poison centers, McFee et al. (35) found that all
recommended a monitoring period at least 6 hours; one rec-
ommended 6–12 hours and two recommended 24 hours. In a
review of TCA poisoning, Callaham (167) suggested that
asymptomatic, decontaminated patients should be monitored
for at least 6 hours (but this would be 6 hours after presenta-
tion to a healthcare facility, not 6 hours after the ingestion).
Later, Callaham and Kassel (3) studied 111 TCA-associated
deaths and concluded that 6 hours is an adequate time to
monitor decontaminated patients for the development of
major signs of toxicity. Others have also recommended a 6-
hour observational period for symptoms after decontamina-
tion measures have been taken (161). Banahan and Schelkun
(168) reviewed 33 cases of TCA overdose (level 4) and con-
cluded that a 6-hour observational period can avert the cost of
unnecessary hospitalization.
Underlying medical conditions/special populations
Patients with underlying heart disease or cardiac arrhyth-
mias or conduction disturbances could be especially sensi-
tive to the toxic effects of TCAs (169). Those with
underlying seizure disorders and taking TCAs could experi-
ence a lowering of the seizure threshold (170). TCAs can
interact with many different medications, including other
psychopharmaceuticals such as MAO inhibitors. It is rea-
sonable to assume that patients taking other drugs (e.g., car-
dioactive agents like digitalis, calcium channel blockers, or
ß-blockers) who overdose on TCAs might have increased
risks of toxicity such that they require immediate triage to a
healthcare facility for monitoring.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
220 A.D. Woolf et al.
Prehospital management of TCA Poisoning
There were few level 1, 2, or 3 articles specifically addressing
out-of-hospital management of TCA exposures (although
many articles contained some limited out-of-hospital infor-
mation). It was felt that some in-hospital data could be uti-
lized to develop out-of-hospital guidelines. Therefore, both
in- and out-of-hospital data are included in this review of the
evidence. While physostigmine, phenytoin, and ß-blockers
have been used to treat TCA poisoned patients in the past,
these agents are not used routinely in the current management
of TCA poisoned patients (19). Moreover, the panel deter-
mined that some hospital-based therapies such as glucagon
and hyperventilation would not be included in this guideline.
Gastrointestinal decontamination
Only decontamination measures that could reasonably be
expected to be available and carried out in a prehospital set-
ting and which had a significant amount of data are reviewed
here.
Ipecac-induced emesis
There were no controlled trials of the use of ipecac syrup for
TCA overdose and no prospective volunteer studies examin-
ing the efficacy of ipecac in reducing TCA absorption even
after therapeutic doses. There were multiple case reports in
which ipecac syrup was administered after suspected TCA
overdoses (68,161,166,171–174). In some of these, tablets or
tablet fragments were recovered (68,166). However, most did
not test the recovered fragments to confirm their identity, nor
did they quantify how much of the ingested dose was recov-
ered. These limitations, coupled with the rare reports of
adverse events as a result of ipecac syrup administration,
make the value of these individual reports questionable.
Activated charcoal
Tricyclic antidepressants are adsorbed by activated charcoal.
Crome et al. (175) demonstrated that 1 g of activated charcoal
adsorbed 318 mg nortriptyline in an in vitro model. Activated
charcoal might be effective in decreasing the absorption of
TCA if given early enough after ingestion.
Investigations of activated charcoal’s effectiveness in stud-
ies performed using TCA poisoned patients have yielded
mixed results. There were several level 2–3 articles in which
activated charcoal was used in an uncontrolled fashion but in
which its efficacy was not reported. In addition, there were
numerous level 4 case reports or series in which single or
multiple doses of activated charcoal were given to individuals
with TCA overdoses, but it was impossible to determine the
efficacy of activated charcoal from these reports given the
lack of controls, the concurrent use of other therapies, the fact
that activated charcoal does not produce immediate clinical
improvement (i.e., outcome is generally measured by
improved pharmacokinetic parameters or the prevention of
later clinical sequelae), and that the interval between the time
of TCA ingestion and administration of charcoal is often
unknown. For many studies of TCA poisoned patients given
activated charcoal in emergency departments, the delay in its
administration for many hours beyond the time of TCA
ingestion might account for an observed lack of effectiveness.
A level 1b, randomized, controlled trial investigated three
decontamination methods in 51 adults with acute unspecified
TCA overdose. There were no significant differences in any
of the outcome measures (clinical and laboratory) between
patients receiving activated charcoal 50 g with magnesium
citrate (group 1), patients receiving gastric lavage followed
by activated charcoal 25 g with magnesium citrate (group 2),
or those receiving activated charcoal 25 g followed by gastric
lavage followed again by activated charcoal (group 3). How-
ever, there were no untreated controls so the effectiveness of
charcoal could not be established (176).
A level 1b, prospective randomized trial in 77 patients
admitted with acute TCA overdose compared the efficacy of
gastric lavage alone vs. gastric lavage plus activated charcoal
20 g. Patients randomized to the activated charcoal group had
no significant differences in peak TCA serum concentration,
drug half-life, or AUC when compared to the gastric lavage-
only patients. Slightly fewer patients in the activated charcoal
group developed convulsions, hypotension, or dysrhythmias
or required prolonged intubation, ICU care, or hospitalization,
but these trends did not reach statistical significance (177).
A level 2b study of 48 children less than 6 years of age
who had been reported to one poison center after acute TCA
exposure found that there was no difference in clinical out-
come between those who had received activated charcoal and
those who had not. However, there were no cases of signifi-
cant toxicity in any of the patients, and some patients had also
received gastric lavage and/or a cathartic (46).
Hedges et al. (178), in a level 2b, prospective study of nine
adults with acute TCA poisoning, detected a correlation
between the time to activated charcoal administration (along
with gastric lavage and supportive care) and reduced serum
half-life of the TCA and an inverse correlation between the
dose of activated charcoal given and drug half-life. In a level
1b study, Crome et al. (179) randomized 48 patients with sus-
pected acute TCA overdose to receive either supportive care
alone or supportive care plus activated charcoal 10 g and
detected no differences in clinical outcome or the rate of fall
of serum concentrations.
In addition to the above studies, there were several articles
that looked at the effect of activated charcoal on TCA absorp-
tion in volunteers given relatively low doses of various anti-
depressants. In a level 1b study, a single 5-g dose of activated
charcoal given 30 minutes after a 75-mg dose of nortriptyline
reduced its mean peak serum concentration by 60% in 12
healthy adult volunteers (175). A level 2b, prospective, cross-
over trial in six adult volunteers who ingested 75 mg nortrip-
tyline compared the efficacy of single- and multiple-dose
activated charcoal in reducing nortriptyline absorption (180).
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 221
Subjects given activated charcoal (amount not reported) at 30
minutes had a 58% mean reduction in peak nortriptyline
serum concentrations and a 55% mean reduction in overall
bioavailability. A similar level 1b, prospective, crossover
trial in six adult volunteers compared the efficacy of activated
charcoal when administered at various times after 75 mg
doses of nortriptyline. Activated charcoal 10 g given at 30
minutes after ingestion resulted in a 77% reduction in mean
peak nortriptyline serum concentration and a 74% reduction
in overall bioavailability; the same dose given at 2 hours
resulted in 37% and 38% reductions, respectively; and, when
given at 4 hours, resulted in reductions of 19% and 13%
(181).
A level 1b, prospective, multi-phase, crossover trial in six
volunteers looked at the effects of various interventions on
amitriptyline pharmacokinetics compared to an untreated
control phase after the ingestion of single doses of 75 mg
amitriptyline. Single doses of 50 g activated charcoal given 5
minutes after amitriptyline prevented the drug’s absorption
(as measured by peak serum concentration, AUC, urinary
excretion, and drug half-life) by 99%. Multiple doses of acti-
vated charcoal beginning at 6 hours after ingestion were
much less effective but still reduced amitriptyline’s peak
serum concentration, half-life, AUC, and urinary excretion
(58). A self-controlled, level 2b trial of doxepin 50 mg given
to eight adult volunteers followed either by a single 15-g dose
of activated charcoal 30 minutes later or multiple doses of
activated charcoal over 24 hours (given at 3, 6, 9, 12, and 24
hours after ingestion) found reduced doxepin AUC after both
interventions compared to controls, but only multiple doses
improved the clearance of doxepin (76). An expert panel con-
vened by the American Academy of Clinical Toxicology and
the European Association of Poison Control Centres & Toxi-
cologists recommended that single-dose charcoal be consid-
ered if a patient has ingested a potentially toxic amount of a
poison up to 1 hour previously, noting that there are insuffi-
cient data to support or exclude its use after 1 hour (182).
Although the prehospital administration of activated char-
coal by emergency medical service personnel has been pro-
posed (183–185), there are no studies of its safety or utility
specifically in TCA poisoning. There are important concerns
about the potential for complications when using activated
charcoal in this context. Instillation of activated charcoal
through an orogastric or nasogastric tube in a comatose
patient can be a source of iatrogenic injury. Godambe et al.
(186) described the case of a 13-year-old girl who, after laps-
ing into coma following a multi-drug overdose including
amitriptyline, had pleuropulmonary activated charcoal instil-
lation from a nasogastric tube, which entered the left main-
stem bronchus and then extended into the parenchyma and
through the visceral pleura. TCA poisoned patients have
aspirated activated charcoal, resulting in severe complications
or death (88). Roy et al. (187) studied 82 consecutive TCA
poisoned patients admitted to an intensive care unit and
found evidence of aspirated activated charcoal in the airways
of 18 of 72 (25%) patients who required intubation, although
their survival rate did not differ from that of those who had
not aspirated (level 4). The authors did not state how many
patients, if any, had received charcoal prior to arrival at the
emergency department.
There are also case reports (level 4) of patients suffering
ileus, toxic megacolon, bowel obstruction, or intestinal
ischemia after TCA poisoning (188,189). In such patients,
administration of any oral medication, including charcoal,
would be contraindicated. Gomez et al. (190) reported a 39-
year-old woman who developed an activated charcoal ster-
colith and intestinal perforation requiring surgery during
treatment of an amitriptyline poisoning. A 21-year-old man
presented with coma and convulsions related to amitriptyline
poisoning, received multiple doses of activated charcoal
(with atropine, lidocaine, defibrillation, gastric lavage, and
other therapies), and developed intestinal obstruction from
inspissated activated charcoal requiring surgery (191).
Other treatment measures
There are many different prehospital treatment measures for
TCA overdose reported in the literature including intubation,
intravenous fluids, and typical supportive measures such as
oxygen, atropine, glucose, naloxone, antiarrhythmics, CPR,
defibrillation or cardioversion, and military anti-shock trou-
sers. Anticonvulsants (e.g., diazepam) could be indicated for
patients who have sustained convulsions or are exhibiting
physical signs of an impending convulsion such as hyperre-
flexia, change in consciousness, tremors, and myoclonus.
None of these have controlled prehospital studies specific to
TCA overdoses. Since they are already routinely used in the
prehospital setting, the anecdotal data are not presented here.
Sodium bicarbonate
There is considerable evidence in both animal and human
studies that alkalinization of the blood can be beneficial in
the management of TCA poisoning. In a rat model, sodium
bicarbonate increased the survival rate of animals poisoned
experimentally with amitriptyline and its benefit was additive
to that of an inotropic agent such as epinephrine or norepi-
nephrine (192). An expert panel convened to consider ACLS
measures after TCA poisoning recommended alkalinization
with hypertonic solutions of sodium bicarbonate (193).
In a 1976 level 4 case series, 11 of 12 children with TCA-
induced arrhthymias responded to intravenous sodium bicar-
bonate with conversions to normal cardiac rhythm (194). In a
level 2b review of 184 patients admitted to an ICU after ami-
triptyline overdose, four of eight patients treated with bicar-
bonate had improvements in cardiac conduction (60). A level
2b, retrospective cohort analysis compared the outcomes of
91 patients with TCA overdose who received bicarbonate
with those of 24 patients who did not. While there was no
direct statistical comparison between treated and untreated
groups, hospital stay was shorter for the treated group. Fur-
thermore, within the bicarbonate group, 20 of 21 patients
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
222 A.D. Woolf et al.
with hypotension had improvements in blood pressure, 39 of
49 patients with QRS prolongations had narrowing of their
QRSs, 40 of 85 patients with altered mental status had
improvements in consciousness, and 42 of 43 patients had
improvements in acidosis. There were no complications from
bicarbonate (195). Urinary acidification with ammonium
chloride or urinary alkalinization with bicarbonate, beginning
1 hour after the ingestion of 75 mg amitriptyline, resulted in a
cumulative excretion of less than 5% of the ingested dose
over 72 hours (58).
There are numerous cases or case series (level 4) in which
patients with TCA overdoses apparently responded favorably
to sodium bicarbonate (improved vital signs, cessation of
dysrhythmias or convulsions, reduction in acidosis, and/or
improved cardiac conduction) in a temporally consistent
manner (22,23,54,89,90,96,149,152,154,158,171,196–214).
In other cases, bicarbonate was given either without improve-
ment or with an unreported response (3,43,82,92,154,173,
174,197,209,212,213,215–223). There were also cases in
which bicarbonate might have resulted in or contributed to
adverse effects (e.g., pulmonary edema or excessive alkale-
mia) (75,82,202). Other reports used other forms of alkaliniz-
ing therapy (e.g., sodium lactate) or unspecified
alkalinization procedures. In some cases these were benefi-
cial but not in others (6,41,44,224).
Intravenous fluids
Intravenous fluids, lidocaine, and respiratory support were all
found to be helpful in stabilizing blood pressure and the car-
diopulmonary status of TCA-poisoned patients in retrospec-
tive level 4 case series (44,225).
Flumazenil
The administration of flumazenil to comatose patients with
unknown poisonings, as both a diagnostic and therapeutic
measure, has occasionally resulted in the unmasking of
convulsions in TCA-poisoned patients who had co-ingested
benzodiazepines (226).
Limitations of the published data
Overall, the level 4 data were difficult to interpret and sum-
marize. The case reports and case series varied widely in the
extent of clinical detail presented, and the cases varied widely
in the severity and clinical effects of poisoning; the timing,
combination, dose, and routes of various treatments used; and
in a number of other patient- or circumstance-specific factors.
Data on the amount ingested were often inaccurate or
incomplete. The history is often obtained from an intoxicated
patient or an emotionally stressed or elderly caregiver. Parents
might underestimate or overestimate an ingested dose because
of denial or anxiety. Poison center personnel often use the
worst-case scenario to estimate an ingested dose in order to
provide a wide margin of safety. In most case reports and case
series, the history of exposure was not independently verified
or confirmed by laboratory testing. Poor correlation between
reported estimated doses and subsequent concentrations or tox-
icity has been documented for children with unintentional
ingestions of other drugs. In most of the cases reviewed, the
exact time of ingestion was not reported or was not known, and
the time of onset of toxicity could only be estimated as occur-
ring within a range of hours after the suspected ingestion.
Conclusions
The expert consensus panel chose to emphasize the impor-
tance of information that would be needed in order to make a
sound triage decision for a patient with tricyclic antidepres-
sant ingestion. These variables include the patient’s intent,
the time of the ingestion, the patient’s symptoms, any under-
lying medical conditions, the dose of the specific product
ingested, and any co-ingested drugs. The expert consensus
panel agreed that in each case, the judgment of the specialist
in poison information or the poison center medical director or
other poison center clinicians might override any specific rec-
ommendation from this guideline.
Patient intent
The expert consensus panel concluded that all patients with
suicidal intention or in whom a malicious intent is suspected
(e.g., child abuse or neglect) should be transported expedi-
tiously by EMS to an emergency department. Patients with-
out these characteristics are candidates for consideration of
prehospital management of their TCA ingestion.
Time since ingestion
The expert consensus panel concluded that decisions about
referral to an emergency department should be based on the
clinical status of the patient within the first 6 hours after TCA
ingestion. The panel concluded that an asymptomatic patient
who unintentionally ingested a TCA is unlikely to develop
symptoms if the interval between the ingestion and the call is
greater than 6 hours. Patients with unintentional TCA poison-
ing who are more than 6 hours after ingestion at the time of
the first contact with a poison center and are still asymptom-
atic can be safely monitored at home.
Patient’s symptoms or underlying medical conditions
The expert consensus panel concluded that referral to an
emergency department should be considered for any patient
who is experiencing symptoms that might be reasonably
related to the TCA (e.g., dizziness, syncope, convulsions,
chest pain, generalized weakness, shortness of breath), who
has severe underlying neurological (e.g., epilepsy) or cardio-
vascular disease (e.g., end-stage cardiomyopathy), or who is
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 223
on multiple psychopharmaceutical or cardiovascular medi-
cines that could have additive neurological or cardiodepres-
sant effects with the TCA. The importance of each of these
variables can be difficult to judge in a telephone conversation
so a low threshold for emergency department evaluation is
considered prudent. Symptomatic patients should be trans-
ported by EMS.
Dose of the drug
The panel concluded that a specific toxic dose of TCA to trigger
referral to an emergency department has limited support of evi-
dence in medical toxicology textbooks, TESS fatality informa-
tion, or the medical literature. The panel recognized the risk of
serious complications after TCA poisoning at relatively low
doses in both children and adults. The panel noted that it is
widely believed among poison centers that older children and
adults are much more likely to have suicidal or homicidal intent.
It concluded that the severity of poisoning that occurred second-
ary to self-harm, intentional misuse, Munchausen-by-proxy,
child abuse, or other malicious intent appeared to result in more
severe outcomes (e.g., fatalities) compared to unintentional
ingestion. This is likely to be a consequence of larger dose and
delayed time to treatment. There is no evidence that age alone
influences the outcome.
The estimation of dose is based largely on the patient’s his-
tory and the type of product and its packaging (when avail-
able for evaluation). If precise data for the ingestion are
unknown or unclear (package size, unit size, number of units
ingested), poison centers in the US often utilize a method in
which the maximum potential dose is calculated.
For asymptomatic patients with acute, unintentional inges-
tions of TCAs, the expert consensus panel concluded that
home observation might be allowable for very small expo-
sures. However, the panel recognized that a definite threshold
dose for toxicity, based on a confirmed history of exposure,
has not been established. After a thorough review of pub-
lished case reports, recommended therapeutic dosage regi-
mens, current poison control center practices, and expert
experience, the panel concluded that ingestion of either of the
following amounts, whichever is lower, should warrant con-
sideration of referral to an emergency department:
An amount that exceeds the usual maximum single thera-
peutic dose; or
An amount equal to or greater than the lowest reported
toxic dose.
Based on this principle, the panel determined that more than
5 mg/kg should be the minimal dose for referral to an emer-
gency department, except in the cases of desipramine (>2.5
mg/kg), nortriptyline (>2.5 mg/kg), trimipramine (>2.5 mg/
kg), and protriptyline (>1 mg/kg). These doses are extrapo-
lated back from the maximum therapeutic adult doses shown
in Table 2. This recommendation applies to both patients who
are naïve to the specific cyclic antidepressant and to patients
currently taking cyclic antidepressants who take extra doses,
in which case the extra doses, when added to the daily dose
taken, should then be compared to the threshold dose for
referral to an ED.
There is support for this threshold dose in retrospective stud-
ies (45–47) that reported little significant toxicity in doses less
than 5 mg/kg, in case reports of minimal doses associated with
sigificant toxicity (Tables 4 and 5), in toxicology textbooks that
often define 10 mg/kg (or 1000 mg) as the dose above which sig-
nificant toxicity is seen, in prospective therapeutic dosing studies
(95,95,112,121,130), and in a meta-analysis in which mean dos-
ing to 3 mg/kg daily of a TCA (2 mg daily for nortriptyline) has
been administered routinely therapeutically to older children and
adults with few significant side effects (106). However, it also
acknowledges recent work showing a comparatively higher case
fatality rate attributable to desipramine exposures compared with
other TCAs, which supports a lower threshold dose of concern
(12,13). This conclusion acknowledges that rare, idiosyncratic
reactions to TCAs, including cardiac arrest, have occurred in
patients taking only therapeutic doses. The panel also concluded
that the toxic dose of doxepin cream preparation, when ingested,
could not be established from the evidence and so chose to
assign the same 5 mg/kg threshold for referral to an emergency
department as oral doxepin dosage forms.
The panel recognized that the decision to send a patient to
an emergency department for monitoring is made on a case-
by-case basis, taking into account the reliability of the
caller’s history, underlying medical status, concomitant use
of other medications (e.g., psychopharmaceutical or cardio-
active agents) that could have additive neurotoxicity or car-
diodepressant effects, and other variables.
Duration of observation for asymptomatic patients
The expert consensus panel concluded that onset and duration
of toxicity were clearly affected by several variables including
the total quantity ingested, co-ingestants, and gastrointestinal
decontamination measures such as activated charcoal. Other
factors, such as type of pharmaceutical product (e.g., capsule
vs. tablet) and the presence or absence of food in the stomach
could also affect the time of onset and duration of toxicity;
however, no studies were found that addressed such differ-
ences directly. After a careful review of the case reports and
observational studies, and considerable discussion, the panel
concluded that a patient with normal sensorium who is asymp-
tomatic 6 hours after ingestion of a TCA is unlikely to subse-
quently develop significant symptoms.
Potential out-of-hospital management
The expert consensus panel concluded that close monitoring
of vital signs, respiratory, cardiovascular, and neurological
status of patients with possible severe TCA poisoning is of
critical importance. When possible, ECG monitoring should
also be included.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
224 A.D. Woolf et al.
Gastrointestinal decontamination
The expert consensus panel concluded that out-of-hospital gas-
trointestinal decontamination offered potential benefit, but that
the magnitude of the benefit and the risks for the patient were
difficult to determine. Inducing emesis with ipecac syrup was
concluded to carry a major risk of pulmonary aspiration of gas-
tric contents if the patient became hypotensive or lost con-
sciousness and is not supported by sufficient evidence of
benefit to warrant its use. Moreover, ipecac syrup would likely
delay or prevent the use of activated charcoal. It might induce a
vagal stimulus that could further depress the heart rate and trig-
ger life-threatening arrhythmias. The panel concluded that ipe-
cac syrup is contraindicated in TCA poisoning.
Activated charcoal was determined by the panel to be a
treatment that could be administered orally as part of the man-
agement of a TCA-poisoned patient, although its effectiveness
and risks have not been evaluated in the prehospital setting.
Aspiration of activated charcoal, with subsequent pulmonary
complications, is a considerable risk of its administration to
TCA-poisoned patients whether in the prehospital or hospital
setting. It cannot be recommended for routine prehospital man-
agement of TCA poisoning at this time, although it might be
considered in some regions in which prehospital activated
charcoal is routinely administered by emergency medical per-
sonnel and there is a long transportation time to an emergency
department. Also, the panel agreed that transportation to an
emergency department should not be delayed in order to
attempt activated charcoal administration.
Specific pharmacological therapy
The expert consensus panel concluded that intravenous flu-
ids would likely be necessary in the prehospital care of
hemodynamically unstable TCA-poisoned patients.
Although the available literature on in-hospital management
of TCA poisoning supports the use of intravenous sodium
bicarbonate, which is often available to paramedics, no
studies were found addressing the effectiveness or safety of
this drug for the out-of-hospital treatment of TCA-induced
hypotension and arrhythmias. However, standard ACLS
doses of sodium bicarbonate can be considered for prehos-
pital patients found to have life-threatening hypotension and
cardiac conduction disturbances evident on an ECG or
rhythm strip. The panel concluded that flumazenil is con-
traindicated if there is any possibility that a comatose
patient may have ingested a TCA.
Recommendations (grades combined
where appropriate)
1. Patients with suspected self-harm or who are the victims
of malicious administration of a TCA should be referred to
an emergency department immediately (Grade D).
2. Patients with acute TCA ingestions who are less than
6 years of age and other patients without evidence of
self-harm should have further evaluation including stan-
dard history taking and determination of the presence of
co-ingestants (especially other psychopharmaceutical
agents) and underlying exacerbating conditions, such as
convulsions or cardiac arrhythmias. Ingestion of a TCA in
combination with other drugs might warrant referral to an
emergency department. The ingestion of a TCA by a
patient with significant underlying cardiovascular or neu-
rological disease should cause referral to an emergency
department at a lower dose than for other individuals.
Because of the potential severity of TCA poisoning, trans-
portation by EMS, with close monitoring of clinical status
and vital signs en route, should be considered (Grade D).
3. Patients who are symptomatic (e.g., weak, drowsy, dizzy,
tremulous, palpitations) after a TCA ingestion should be
referred to an emergency department (Grade B).
4. Ingestion of either of the following amounts (whichever is
lower) would warrant consideration of referral to an emer-
gency department:
An amount that exceeds the usual maximum single
therapeutic dose or,
An amount equal to or greater than the lowest reported
toxic dose.
For all TCAs except desipramine, nortriptyline, trimipramine,
and protriptyline, this dose is >5 mg/kg. For despiramine it is
>2.5 mg/kg; and for nortriptyline it is >2.5 mg/kg; for trimi-
pramine it is >2.5 mg/kg; for protriptyline it is >1 mg/kg. This
recommendation applies to both patients who are naïve to the
specific cyclic antidepressant and to patients currently taking
cyclic antidepressants who take extra doses, in which case the
extra doses should be added to the daily dose taken and then
compared to the threshold dose for referral to an emergency
department (Grades B/C).
5. Do not induce emesis (Grade D).
6. The risk-to-benefit ratio of prehospital activated charcoal for
gastrointestinal decontamination in TCA poisoning is unknown.
Prehospital activated charcoal administration, if available,
should only be carried out by health professionals and only if no
contraindications are present. Do not delay transportation in
order to administer activated charcoal (Grades B/D).
7. For unintentional poisonings, asymptomatic patients are
unlikely to develop symptoms if the interval between the
ingestion and the initial call to a poison center is greater
than 6 hours. These patients do not need referral to an
emergency department facility (Grade C).
8. Follow-up calls to determine the outcome for a TCA
ingestions ideally should be made within 4 hours of the
initial call to a poison center and then at appropriate
intervals thereafter based on the clinical judgment of the
poison center staff (Grade D).
9. An ECG or rhythm strip, if available, should be checked
during the prehospital assessment of a TCA overdose
patient. A wide-complex arrhythmia with a QRS duration
longer than 100 msec is an indicator that the patient
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 225
should be immediately stabilized, given sodium bicar-
bonate if there is a protocol for its use, and transported to
an emergency department (Grade B).
10. Symptomatic patients with TCA poisoning might require
prehospital interventions, such as intravenous fluids, car-
diovascular agents, and respiratory support, in accor-
dance with standard ACLS guidelines as outlined by the
American Heart Association (227) (Grade D).
11. Administration of sodium bicarbonate might be benefi-
cial for patients with severe or life-threatening TCA tox-
icity if there is a prehospital protocol for its use (Grades
B/D).
12. For TCA-associated convulsions, benzodiazepines are
recommended (Grade D).
13. Flumazenil is not recommended for patients with TCA
poisoning (Grade D).
Dosage and follow-up recommendations are summarized in
Appendix 4.
Implications for research
The expert consensus panel identified the following topics
where additional research might be useful.
1. A prospective study of the dose of acute pediatric TCA inges-
tion requiring observation at a healthcare facility could help
to reduce unnecessary utilization of healthcare resources.
2. A large-scale, prospective study of unintentional TCA
ingestions is needed.
3. An additional need is a better correlation between the esti-
mated ingested dose, clinical symptoms, and outcome in
patients with serious overdoses.
4. Research is needed into the adverse effects of TCAs in
pregnant women.
5. The efficacy and safety of prehospital decontamination
with activated charcoal for TCA poisoning is unknown
and merits investigation.
6. The efficacy and safety of glucagon, sodium bicarbonate,
or other measures given in a prehospital setting for the
treatment of TCA poisoning is unknown.
Disclosures
Dr. Booze’s husband is employed by AstraZeneca. Dr. Erdman
is currently employed by Genentech but was not during his
contribution to the development of this guideline. There are
no other potential conflicts of interest reported by the expert
consensus panel or project staff regarding this guideline.
References
1. Watson WA, Litovitz TL, Rodgers GC, Klein-Schwartz W, Reid N,
Youniss J, Flanagan A, Wruk KM. 2004 annual report of the American
Association of Poison Control Centers Toxic Exposure Surveillance
System. Am J Emerg Med 2005; 23:589–666.
2. Freeman JW, Mundy GR, Beattie RR, Ryan C. Cardiac abnormalities in
poisoning with tricyclic antidepressants. Br Med J 1969; 2:610–611.
3. Callaham M, Kassel D. Epidemiology of fatal tricyclic antidepressant
ingestion: implications for management. Ann Emerg Med 1985; 14:1–9.
4. Jick SS, Dean AD, Jick H. Antidepressants and suicide. BMJ 1995;
310:215–218.
5. Olson KR, Kearney TE, Dyer JE, Benowitz NL, Blanc PD. Seizures
associated with poisoning and drug overdose. Am J Emerg Med 1993;
11:565–568.
6. Taboulet P, Michard F, Muszynski J, Galliot-Guilley M, Bismuth C.
Cardiovascular repercussions of seizures during cyclic antidepressant
poisoning. J Toxicol Clin Toxicol 1995; 33:205–211.
7. Crome P, Newman B. Fatal tricyclic antidepressant poisoning. J R Soc
Med 1979; 72:649–653.
8. Henry JA. A fatal toxicity index for antidepressant poisoning. Acta Psy-
chiatr Scand Suppl 1989; 354:37–45.
9. Henry JA, Alexander CA, Sener EK. Relative mortality from overdose
of antidepressants. BMJ 1995; 310:221–224.
10. Farmer RD, Pinder RM. Why do fatal overdose rates vary between anti-
depressants? Acta Psychiatr Scand Suppl 1989; 354:25–35.
11. Wedin GP, Oderda GM, Klein-Schwartz W, Gorman RL. Relative tox-
icity of cyclic antidepressants. Ann Emerg Med 1986; 15:797–804.
12. Amitai Y, Frischer H. Excess fatality from desipramine and dosage rec-
ommendations. Ther Drug Monit 2004; 26:468–473.
13. Amitai Y, Frischer H. Excess fatality from desipramine in children and
adolescents. J Am Acad Child Adolesc Psychiatry 2006; 45:54–60.
14. Baldessarini RJ. Drugs and the treatment of psychiatric disorders:
depression and anxiety disorders. In: Hardman JG, Limbird LE, eds.
Goodman & Gilman’s the Pharmacological Basis of Therapeutics. 10th
ed. New York: McGraw-Hill, 2001:447–483.
15. Cohen LG, Biederman J, Wilens TE, Spencer TJ, Mick E, Faraone SV,
Prince J, Flood JG. Desipramine clearance in children and adolescents:
absence of effect of development and gender. J Am Acad Child Adolesc
Psychiatry 1999; 38:79–85.
16. Swartz CM, Sherman A. The treatment of tricyclic antidepressant over-
dose with repeated charcoal. J Clin Psychopharmacol 1984; 4:336–340.
17. Rauber A, Maroncelli R. Prescribing practices and knowledge of tricy-
clic antidepressants among physicians caring for children. Pediatrics
1984; 73:107–109.
18. Brown-Cartwright D, Brater DC, Barnett CC, Richardson CT. Effect of
doxepin on basal gastric acid and salivary secretion in patients with
duodenal ulcer. Ann Intern Med 1986; 104:204–206.
19. Klasco RK, ed. Poisindex system. Greenwood Village (CO): Thomson
Micromedex, edition expires March 2004.
20. Miadinich EK, Carlow TJ. Total gaze paresis in amitriptyline overdose.
Neurology 1977; 27:695.
21. Smith MS. Amitriptyline ophthalmoplegia. Ann Intern Med 1979;
91:793.
22. Roberge RJ, Martin TG, Hodgman M, Benitez JG. Acute chemical pan-
creatitis associated with a tricyclic antidepressant (clomipramine) over-
dose. J Toxicol Clin Toxicol 1994; 32:425–429.
23. Mullins ME, Cristofani CB, Warden CR, Cleary JF. Amitriptyline-asso-
ciated seizures in a toddler with Munchausen-by-proxy. Pediatr Emerg
Care 1999; 15:202–205.
24. Winrow AP. Amitriptyline-associated seizures in a toddler with
Munchausen-by-proxy. Pediatr Emerg Care 1999; 15:462–463.
25. Simon FA, Treuting JJ. Nonaccidental poisoning in a two-month-old
child. Clin Toxicol 1981; 18:37–40.
26. Watson JB, Davies JM, Hunter JL. Nonaccidental poisoning in child-
hood. Arch Dis Child 1979; 54:143–144.
27. Shaneyfelt TM, Mayo-Smith MF, Rothwangl J. Are guidelines following
guidelines? The methodological quality of clinical practice guidelines in
the peer-reviewed medical literature. JAMA 1999; 281:1900–1905.
28. Shiffman RN, Shekelle P, Overhage JM, Slutsky J, Grimshaw J,
Deshpande AM. Standardized reporting of clinical practice guidelines:
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
226 A.D. Woolf et al.
a proposal from the Conference on Guideline Standardization. Ann
Intern Med 2003; 139:493–498.
29. Benowitz N. Antidepressants, tricyclic. In: Olson KR, ed. Poisoning &
Drug Overdose. 4th ed. New York: McGraw-Hill, 2004:90–93.
30. Dawson AH. Cyclic antidepressants. In: Dart RC, ed. Medical Toxicology.
3rd ed. Philadelphia: Lippincott Williams & Wilkins, 2004:852–861.
31. Geis GL, Bond GR. Antidepressant overdose: tricyclics, selective sero-
tonin reputake inhibitors, and atypical antidepressants. In: Erickson TR,
Ahrens WR, Aks SE, Baum CR, Ling LJ, eds. Pediatric Toxicology:
Diagnosis & Management of the Poisoned Child. New York: McGraw-
Hill, 2005:297–302.
32. Liebelt EL, Francis PD. Cyclic antidepressants. In: Goldfrank LR,
Howland MA, Flomenbaum NE, Hoffman RS, Lewin NA, Nelson LS,
eds. Goldfrank’s Toxicologic Emergencies, 7th ed. New York:
McGraw-Hill, 2002:847–864.
33. Pentel PR, Keyler DE, Haddad LM. Tricyclic antidepressants and selec-
tive serotonin reuptake inhibitors. In: Haddad LM, Shannon MW, Win-
chester JF. Clinical Management of Poisoning and Drug Overdose. 3rd
ed. Philadelphia: WB Saunders, 1998:437–451.
34. Kuczmarski RJ, Ogden CL, Grummer-Strawn LM, Flegal KM, Guo SS,
Wei R, Mei Z, Curtin LR, Roche AF, Johnson CL. CDC growth charts:
Unied States. Advance data from vital and health statistics; no. 314.
Hyattsville, MD; National Center for Health Statistics, 2004. Available
at: http://www.cdc.gov/nchs/data/ad/ad314.pdf
35. McFee RB, Mofenson HC, Caraccio TR. A nationwide survey of the man-
agement of unintentional-low dose tricyclic antidepressant ingestions
involving asymptomatic children: implications for the development of an
evidence-based clinical guideline. J Toxicol Clin Toxicol 2000; 38:15–19.
36. McFee RB, Mofenson HC, Caraccio TR. A nationwide survey of poi-
son control centers comparing 1999 to 1998 triage and management of
asymptomatic children who ingested tricyclic antidepressant. Vet Hum
Toxicol 2001; 43:305–307.
37. Steel CM, O’Duffy J, Brown SS. Clinical effects and treatment of imi-
pramine and amitriptyline poisoning in children. Br Med J 1967;
3:663–667.
38. Goel KM, Shanks RA. Amitriptyline and imipramine poisoning in chil-
dren. Br Med J 1974; 1:261–263.
39. Berkovitch M, Matsui D, Fogelman R, Komar L, Hamilton R, Johnson
D. Assessment of the terminal 40-millisecond QRS vector in children
with a history of tricyclic antidepressant ingestion. Pediatr Emerg Care
1995; 11:75–77.
40. Biggs JT, Spiker DG, Petit JM, Ziegler VE. Tricyclic antidepressant
overdose: incidence of symptoms. JAMA 1977; 238:135–138.
41. Bouffard Y, Palmier B, Bouletreau P, Motin J. Intoxication aiguë par
les antidépresseurs tricycliques. Critères de gravité et traitement. Étude
de 16 observations avec manifestations cardio-vasculaires. Ann Med
Interne (Paris) 1982; 133:256–260.
42. Brown TC, Dwyer ME, Stocks JG. Antidepressant overdosage in chil-
dren--a new menace. Med J Aust 1971; 2:848–851.
43. Christensen KN, Andersen HH. Deliberate poisoning with tricyclic
antidepressants treated in an intensive care unit. Acta Pharmacol Toxi-
col (Copenh) 1977; 41(Suppl 2):511–515.
44. deCastro FJ, Rost K, Jaeger R. Tricyclic antidepressant poisoning: clin-
ical series. Vet Hum Toxicol 1980; 22:68–69.
45. McFee RB, Caraccio TR, Mofenson HC. Tricyclic antidepressant
(TCA) ingestions: pediatric toxicological management considerations
by a regional poison control center (RPCC) [abstract]. J Toxicol Clin
Toxicol 1998; 36:518.
46. McFee RB, Caraccio TR, Mofenson HC. Selected tricyclic antidepres-
sant ingestions involving children 6 years old or less. Acad Emerg Med
2001; 8:139–144.
47. Spiller HA, Baker SD, Krenzelok EP, Cutino L. Use of dosage as a tri-
age guideline for unintentional cyclic antidepressant (UCA) ingestions
in children. Am J Emerg Med 2003; 21:422–424.
48. Strom J, Sloth Madsen P, Nygaard Nielsen N, Bredgaard Sorensen M.
Acute self-poisoning with tricyclic antidepressants in 295 consecutive
patients treated in an ICU. Acta Anaesthesiol Scand 1984; 28:666–670.
49. Jue SG. Desipramine--accidental poisoning. Drug Intell Clin Pharm
1976; 10:52–53.
50. Giles HM. Imipramine poisoning in childhood. Br Med J 1963;
5361:844–846.
51. Serafimovski N, Thorball N, Asmussen I, Lunding M. Tricyclic antide-
pressive poisoning with special reference to cardiac complications.
Acta Anaesthesiol Scand Suppl 1975; 57:55–63.
52. Chamsi-Pasha H, Barnes PC. Myocardial infarction: a complication of
amitriptyline overdose. Postgrad Med J 1988; 64:968–970.
53. Bramble MG, Lishman AH, Purdon J, Diffey BL, Hall RJ. An analysis
of plasma levels and 24-hour ECG recordings in tricyclic antidepressant
poisoning: implications for management. Q J Med 1985; 56:357–366.
54. Ellison DW, Pentel PR. Clinical features and consequences of seizures
due to cyclic antidepressant overdose. Am J Emerg Med 1989; 7:5–10.
55. Noto R, Robert J, Hanote P. Traitement d’urgence et transport des
intoxications aiguës par les dérivés imipraminiques (à propos de 70
cas). Agressologie 1970; 11:515–523.
56. Pellinen TJ, Farkkila M, Heikkila J, Luomanmaki K. Electrocardio-
graphic and clinical features of tricyclic antidepressant intoxication. A
survey of 88 cases and outlines of therapy. Ann Clin Res 1987; 19:12–17.
57. Bye C, Clubley M, Peck AW. Drowsiness, impaired performance and
tricyclic antidepressant drugs. Br J Clin Pharmacol 1978; 6:155–161.
58. Karkkainen S, Neuvonen PJ. Pharmacokinetics of amitriptyline influ-
enced by oral charcoal and urine pH. Int J Clin Pharmacol 1986;
24:326–332.
59. Hulten BA, Heath A, Knudsen K, Nyberg G, Starmark JE, Martensson
E. Severe amitriptyline overdose: relationship between toxicokinetics
and toxicodynamics. J Toxicol Clin Toxicol 1992; 30:171–179.
60. Köppel C, Wiegreffe A, Tenczer J. Clinical course, therapy, outcome
and analytical data in amitriptyline and combined amitriptyline/chlor-
diazepoxide overdose. Hum Exp Toxicol 1992; 11:458–465.
61. Rudorfer MV, Robins E. Amitriptyline overdose: clinical effects on
tricyclic antidepressant plasma levels. J Clin Psychiatry 1982;
43:457–460.
62. Siddiqui JH, Vakassi MM, Ghani MF. Cardiac effects of amitriptyline
overdosage. Curr Ther Res Clin Exp 1977; 22:321–325.
63. Aquilonius SM, Hedstrand U. The use of physostigmine as an antidote in tri-
cyclic anti-depressant intoxication. Acta Anaesthesiol Scand 1978; 22:40–45.
64. Pall H, Czech K, Kotzaurek R, Kleinberger G, Pichler M. Experiences
with physostigminesalicylate in tricyclic antidepressant poisoning. Acta
Pharmacol Toxicol (Copenh) 1977; 41:171–178.
65. Vohra J, Burrows G, Hunt D, Sloman G. The effect of toxic and thera-
peutic doses of tricyclic antidepressant drugs on intracardiac conduc-
tion. Eur J Cardiol 1975; 3:219–227.
66. Dale O, Hole A. Biphasic time-course of serum concentrations of clo-
mipramine and desmethylclomipramine after a near-fatal overdose. Vet
Hum Toxicol 1994; 36:309–310.
67. Bucher HW, Stucki P. Kardial komplikationen bei einer vergiftung mit
desipramine (Pertofran). Schweiz Med Wochenschr 1967; 97:519–521.
68. Callaham M. Admission criteria for tricyclic antidepressant ingestion.
West J Med 1982; 137:425–429.
69. Chahine RA, Castellanos A, Jr. Myocardial toxicity produced by
desipramine overdosage. Chest 1971; 59:566–568.
70. Colvard C, Jr. Overdosage of desipramine hydrochloride with marked
electrocardiographic abnormalities. South Med J 1968; 61:1218 passim.
71. Hagerman GA, Hanashiro PK. Reversal of tricyclic-antidepressant-
induced cardiac conduction abnormalities by phenytoin. Ann Emerg
Med 1981; 10:82–86.
72. Lee WR, Sheikh MU, Covarrubias EA, Slotkoff LM. Variant ven-
tricular tachycardia in desipramine toxicity. South Med J 1981;
74:1268–1269.
73. Shannon M. Toxicology reviews: physostigmine. Pediatr Emerg Care
1998; 14:224–226.
74. Williams AJ. “Desipramine” overdosage. Br Med J 1964; 1:371–372.
75. Zuckerman GB, Conway EE, Jr. Pulmonary complications following
tricyclic antidepressant overdose in an adolescent. Ann Pharmacother
1993; 27:572–574.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 227
76. Scheinin M, Virtanen R, Iisalo E. Effect of single and repeated doses of
activated charcoal on the pharmacokinetics of doxepin. Int J Clin Phar-
macol 1985; 23:38–42.
77. Bastani JR. Physostigmine treatment of tricyclic overdosage in psycho-
sis. Psychosomatics 1979; 20:847–848.
78. Cordonnier J, Heyndrickx A, Jordaens L, Brijs R, De Keyser R. A fatal
intoxication due to doxepin. J Anal Toxicol 1983; 7:161–164.
79. Janson PA, Watt JB, Hermos JA. Doxepin overdose: success with phys-
ostigmine and failure with neostigmine in reversing toxicity. JAMA
1977; 237:2632–2633.
80. Oliver JS, Watson AA. Doxepin poisoning. Med Sci Law 1974;
14:280–283.
81. Williams JO. Respiratory depression in tricyclic overdose. Br Med J
1972; 1:631.
82. Wrenn K, Smith BA, Slovis CM. Profound alkalemia during treatment
of tricyclic antidepressant overdose: a potential hazard of combined
hyperventilation and intravenous bicarbonate. Am J Emerg Med 1992;
10:553–555.
83. Goldberg MJ, Park GD, Spector R, Fischer LJ, Feldman RD. Lack of
effect of oral activated charcoal on imipramine clearance. Clin Pharma-
col Ther 1985; 38:350–353.
84. Vohra J, Hunt D, Burrows G, Sloman G. Intracardiac conduction
defects following overdose of tricyclic antidepressant drugs. Eur J Car-
diol 1975; 2:443–452.
85. Stinnett JL, Valentine J, Abrutyn E. Nortriptyline hydrochloride over-
dosage. JAMA 1968; 204:69–71.
86. Brackenridge RG, Peters TJ, Watson JM. Myocardial damage in ami-
triptyline and nortriptyline poisoning. Scott Med J 1968; 13:208–210.
87. Duke WW, Horton JP. Nortriptyline (Aventyl) overdosage. South Med
J 1969; 62:1348–1349.
88. Elliott CG, Colby TV, Kelly TM, Hicks HG. Charcoal lung. Bron-
chiolitis obliterans after aspiration of activated charcoal. Chest
1989; 96:672–674.
89. Lipper B, Bell A, Gaynor B. Recurrent hypotension immediately after
seizures in nortriptyline overdose. Am J Emerg Med 1994; 12:452–453.
90. McKinney PE, Rasmussen R. Reversal of severe tricyclic antidepres-
sant-induced cardiotoxicity with intravenous hypertonic saline solution.
Ann Emerg Med 2003; 42:20–24.
91. Rudorfer MV, Robins E. Fatal nortriptyline overdose, plasma levels,
and in vivo methylation of tricyclic antidepressants. Am J Psychiatry
1981; 138:982–983.
92. Sedal L, Korman MG, Williams PO, Mushin G. Overdosage of tricyclic
antidepressants. A report of two deaths and a prospective study of 24
patients. Med J Aust 1972; 2:74–79.
93. Chin LS, Havill JH, Rothwell RP, Bishop BG. Use of physostigmine
in tricyclic antidepressant poisoning. Anaesth Intensive Care 1976;
4:138–140.
94. Forsythe WI, Merrett JD, Redmond A. Controlled clinical trial of trimi-
pramine and placebo in the treatment of enuresis. Br J Clin Pract 1972;
26:119–121.
95. Bartels MG, Varley CK, Mitchell J, Stamm SJ. Pediatric cardiovascular
effects of imipramine and desipramine. J Am Acad Child Adolesc Psy-
chiatry 1991; 30:100–103.
96. Brown D, Winsberg BG, Bailer I, Press M. Imipramine therapy and
seizures: three children treated for hyperactive behavioral disorders.
Am J Psychiatry 1973; 130:210–212.
97. Petti TA, Campbell M. Imipramine and seizures. Am J Psychiatry
1975; 132:538–540.
98. Leonard HL, Meyer MC, Swedo SE, Richter D, Hamburger SD, Allen
AJ, Rapoport JL, Tucker E. Electrocardiographic changes during
desipramine and clomipramine treatment in children and adolescents.
J Am Acad Child Adolesc Psychiat 1995; 34: 1460–1468.
99. Glassman AH, Giardina EV, Perel JM, Bigger JT, Kantor SJ, Perel
JM, Davies M.. Clinical characteristics of imipramine induced orthos-
tatic hypotension. Lancet 1979; 1:468–472.
100. Wilens TE, Biederman J, Baldessarini RJ, Geller B, Schleifer D, Spen-
cer TJ, Birmaher B, Goldblatt A. Cardiovascular effects of therapeutic
doses of tricyclic antidepressants in children and adolescents. J Am
Acad Child Adolesc Psychiatry 1996; 35:1491–1501.
101. Biederman J. Sudden death in children treated with a tricyclic
antidepressant. J Am Acad Child Adolesc Psychiatry 1991;
30:495–498.
102. Biederman J, Baldessarini RJ, Goldblatt A, Lapey KA, Doyle A,
Hesslein PS. A naturalistic study of 24-hour electrocardiographic
recordings and echocardiographic findings in children and adolescents
treated with desipramine. J Am Acad Child Adolesc Psychiatry 1993;
32:805–813.
103. Miescke KJ, Musa MN. On mixtures of three normal populations
caused by monogenic inheritance: application to desipramine metabo-
lism.J Psychiat Neurosci 1994; 19:295–300.
104. Musa MN, Miescke KJ. Pharmacogenetics of desipramine metabo-
lism. Int J Clin Pharmacol Ther 1994; 32:126–130.
105. Varley CK, McClellan J. Case study: two additional sudden deaths
with tricyclic antidepressants. J Am Acad Child Adolesc Psychiatry
1997; 36:390–394.
106. Wilens TE, Stern TA, O’Gara PT. Adverse cardiac effects of com-
bined neuroleptic ingestion and tricyclic antidepressant overdose. J
Clin Psychopharmacol 1990; 10:51–54.
107. Kaumeier HS, Haase HJ. A double-blind comparison between amox-
apine and amitriptyline in depressed in-patients. Int J Clin Pharmacol
1980; 18:177–184.
108. Veith RC, Bloom V, Bielski R, Friedel RO. ECG effects of compara-
ble plasma concentrations of desipramine and amitriptyline. J Clin
Psychopharmacol 1982; 2:394–398.
109. Burckhardt D, Raeder E, Muller V, Imhof P, Neubauer H. Cardiovas-
cular effects of tricyclic and tetracyclic antidepressa3nts. JAMA 1978;
239:213–216.
110. Milner G, Buckler EG. Adynamic ileus and amitriptyline: three case
reports. Med J Aust 1964; 14:921–922.
111. Rechlin T. Decreased R-R variation: a criterium for overdosage of tri-
cyclic psychotropic drugs. Intens Care Med 1995; 21:598–601.
112. Vohra J, Burrows GD, Sloman G. Assessment of cardiovascular side
effects of therapeutic doses of tricyclic antidepressant drugs. Aust N Z
J Med 1975; 5:7–11.
113. Burkitt EA, Sutcliffe CK. Paralytic ileus after amitriptyline (“Trypti-
zol”). Brit Med J 1961; 1648–1649.
114. Goldsmith HJ. Amitriptyline poisoning. Lancet 1965; 2:640–641.
115. Giller EL, Jr., Bialos DS, Docherty JP, Jatlow P, Harkness L. Chronic
amitriptyline toxicity. Am J Psychiatry 1979; 136:458–459.
116. Heiser JF, Wilbert DE. Reversal of delirium induced by tricyclic anti-
depressant drugs with physostigmine. Am J Psychiatry 1974;
131:1275–1277.
117. Johnson PB. Physostigmine in tricyclic antidepressant overdose.
JACEP 1976; 5:443–445.
118. Brasic JR, Barnett JY, Sheitman BB, Tsaltas MO. Adverse effects
of clomipramine. J Am Acad Child Adolesc Psychiatry 1997;
36:1165–1166.
119. Schubert DSP, Miller SI. Are divided doses of tricyclic antidepres-
sants necessary? J Nerv Ment Dis 1978; 166:875–877.
120. Rapoport JL, Mikkelsen EJ, Zavadil A, Nee L, Gruenau C, Mendelson
W, Gillin JC. Childhood enuresis. II. Psychopathology, tricyclic
concentration in plasma, and antienuretic effect. Arch Gen Psychiatry
1980; 37:1146–1152.
121. Schroeder JS, Mullin AV, Elliott GR, Steiner H, Nichols M, Gordon
A, Paulos M. Cardiovascular effects of desipramine in children. J Am
Acad Child Adolesc Psychiatry 1989; 28:376–379.
122. Preskorn SH, Fast GA. Tricyclic antidepressant-induced seizures and
plasma drug concentration. J Clin Psychiatry 1992; 53:160–162.
123. Riddle MA, Nelson JC, Kleinman CS, Rasmusson A, Leckman JF,
King RA, Cohen DJ. Sudden death in children receiving Norpramin: a
review of three reported cases and commentary. J Am Acad Child
Adolesc Psychiatry 1991; 30:104–108.
124. Wagner KD, Fershtman M. Potential mechanism of desipramine-
related sudden death in children. Psychosomatics 1993; 34:80–83.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
228 A.D. Woolf et al.
125. Varley CK. Sudden death related to selected tricyclic antidepressants
in children: epidemiology, mechanisms and clinical implications. Pae-
diatr Drugs 2001; 3:613–627.
126. Riddle MA, Geller B, Ryan N. Another sudden death in a child treated with
desipramine. J Am Acad Child Adolesc Psychiatry 1993; 32:792–797.
127. Fritz GK, Rockney RM, Yeung AS. Plasma levels and efficacy of imi-
pramine treatment for enuresis. J Am Acad Child Adolesc Psychia-
tryry 1994; 33:60–64.
128. Saraf KR, Klein DF, Gittelman-Klein R, Groff S. Imipramine side
effects in children. Psychopharmacologia 1974; 37:265–274.
129. Azima H, Vispo RH. Imipramine; a potent new anti-depressant com-
pound. Am J Psychiatry 1958; 115:245–246.
130. Fletcher SE, Case CL, Sallee FR, Hand LD, Gillette PC. Prospective
study of the electrocardiographic effects of imipramine in children. J
Pediatr 1993; 122:652–654.
131. Winsberg BG, Goldstein S, Yepes LE, Perel JM. Imipramine and elec-
trocardiographic abnormalities in hyperactive children. Am J Psychia-
try 1975; 132:542–545.
132. Saraf K, Klein DF. The safety of a single daily dose schedule for imi-
pramine. Am J Psychiatry 1971; 128:483–484.
133. Brooke G, Weatherly JRC. Imipramine. Lancet 1959; 2:568–569.
134. English D. Balanced treatment of depression. Curr Ther Res Clin Exp
1959; 1:135–138.
135. Mann AM, Catterson AG, Macpherson AS. Toxicity of imipramine:
report on serious side effects and massive overdosage. Can Med Assoc
J 1959; 81:23–28.
136. Levene LJ, Lascelles CF. Imipramine. Lancet 1959; 2:675.
137. Moccetti T, Lichtlen P, Albert H, Meier E, Imbach P. Kardiotoxizität der
trizyklischen antidepressiva. Schweiz Med Wochenschr 1971; 101:1–10.
138. Williams RB, Sherter C. Cardiac complications of tricyclic antidepres-
sant therapy. Ann Intern Med 1971; 74:395–398.
139. Reed K, Smith RC, Schoolar JC, Hu R, Leelavathi DE, Mann E, Lipp-
man L. Cardiovascular effects of nortriptyline in geriatric patients. Am
J Psychiatry 1980; 137:986–988.
140. Ziegler VE, Co BT, Biggs JT. Plasma nortriptyline levels and ECG
findings. Am J Psychiatry 1977; 134:441–443.
141. Schneider LS. QRS duration in acute overdose of tricyclic antidepres-
sants. N Engl J Med 1986; 314:989.
142. Unrecognized, excessive dose of nortriptyline. Int Pharm J 1998;
12:145–146.
143. Hulten BA, Heath A. Clinical aspects of tricyclic antidepressant poi-
soning. Acta Med Scand 1983; 213:275–278.
144. Boehnert MT, Lovejoy FH, Jr. Value of the QRS duration versus the
serum drug level in predicting seizures and ventricular arrhythmias
after an acute overdose of tricyclic antidepressants. N Engl J Med
1985; 313:474–479.
145. Niemann JT, Bessen HA, Rothstein RJ, Laks MM. Electrocardio-
graphic criteria for tricyclic antidepressant cardiotoxicity. Am J Car-
diol 1986; 57:1154–1159.
146. Wolfe TR, Caravati EM, Rollins DE. Terminal 40-ms frontal plane
QRS axis as a marker for tricyclic antidepressant overdose. Ann
Emerg Med 1989; 18:348–351.
147. Liebelt EL, Francis PD, Woolf AD. ECG lead aVR versus QRS inter-
val in predicting seizures and arrhythmias in acute tricyclic antidepres-
sant toxicity. Ann Emerg Med 1995; 26:195–201.
148. Bailey B, Buckley NA, Amre DK. A meta-analysis of prognostic indi-
cators to predict seizures, arrhythmias or death after tricyclic antide-
pressant overdose. J Toxicol Clin Toxicol 2004; 42:877–888.
149. Buckley BM, Boldy DA, Vale JA. The importance of pH and blood
gas monitoring after overdoses of tricyclic antidepressants. Br Med J
(Clin Res Ed) 1984; 289:185.
150. Cronin AJ, Khalil R, Little TM. Poisoning with tricyclic antidepres-
sants: an avoidable cause of childhood deaths. Br Med J 1979; 1:722.
151. Freeman JW, Loughhead MG. Beta blockade in the treatment of tricy-
clic antidepressant overdosage. Med J Aust 1973; 1:1233–1235.
152. Harrigan RA, Brady WJ. ECG abnormalities in tricyclic antidepres-
sant ingestion. Am J Emerg Med 1999; 17:387–393.
153. Masters AB. Delayed death in imipramine poisoning. Br Med J 1967;
3:866–867.
154. McAlpine SB, Calabro JJ, Robinson MD, Burkle FM, Jr. Late death in
tricyclic antidepressant overdose revisited. Ann Emerg Med 1986;
15:1349–1352.
155. McMahon AJ. Amitriptyline overdose complicated by intestinal
pseudo-obstruction and caecal perforation. Postgrad Med J 1989;
65:948–949.
156. Foulke GE, Albertson TE, Walby WF. Tricyclic antidepressant over-
dose: emergency department findings as predictors of clinical course.
Am J Emerg Med 1986; 4:496–500.
157. Hulten BA, Adams R, Askenasi R, Dallos V, Dawling S, Volans G,
Heath A. Predicting severity of tricyclic antidepressant overdose. J
Toxicol Clin Toxicol 1992; 30:161–170.
158. Starkey IR, Lawson AA. Poisoning with tricyclic and related antide-
pressants--a ten-year review. Q J Med 1980; 49:33–49.
159. Greenland P, Howe TA. Cardiac monitoring in tricyclic antidepressant
overdose. Heart Lung 1981; 10:856–859.
160. Petit JM, Spiker DG, Ruwitch JF, Ziegler VE, Weiss AN, Biggs JT.
Tricyclic antidepressant plasma levels and adverse effects after over-
dose. Clin Pharmacol Ther 1977; 21:47–51.
161. Tokarski GF, Young MJ. Criteria for admitting patients with tricyclic
antidepressant overdose. J Emerg Med 1988; 6:121–124.
162. Stern TA, O’Gara PT, Mulley AG, Singer DE, Thibault GE. Compli-
cations after overdose with tricyclic antidepressants. Crit Care Med
1985; 13:672–674.
163. Fasoli RA, Glauser FL. Cardiac arrhythmias and ECG abnormalities in
tricyclic antidepressant overdose. Clin Toxicol 1981; 18:155–163.
164. Emerman CL, Connors AF, Jr., Burma GM. Level of consciousness as
a predictor of complications following tricyclic overdose. Ann Emerg
Med 1987; 16:326–330.
165. Foulke GE. Identifying toxicity risk early after antidepressant over-
dose. Am J Emerg Med 1995; 13:123–126.
166. Pentel P, Sioris L. Incidence of late arrhythmias following tricyclic
antidepressant overdose. Clin Toxicol 1981; 18:543–548.
167. Callaham M. Tricyclic antidepressant overdose. JACEP 1979;
8:413–425.
168. Banahan BF, Jr., Schelkun PH. Tricyclic antidepressant overdose:
conservative management in a community hospital with cost-saving
implications. J Emerg Med 1990; 8:451–454.
169. Glassman AH, Johnson LL, Giardina EG, Walsh BT, Roose SP, Coo-
per TB, Bigger JT Jr. The use of imipramine in depressed patients with
congestive heart failure. JAMA 1983; 250:1997–2001.
170. Rosenstein DL, Nelson JC, Jacobs SC. Seizures associated with anti-
depressants: A review. J Clin Psychiatry 1993; 54:289–299.
171. Molloy DW, Penner SB, Rabson J, Hall KW. Use of sodium bicarbon-
ate to treat tricyclic antidepressant-induced arrhythmias in a patient
with alkalosis. Can Med Assoc J 1984; 130:1457–1459.
172. Phillips S, Brent J, Kulig K, Heiligenstein J, Birkett M. Fluoxetine
versus tricyclic antidepressants: a prospective multicenter study of
antidepressant drug overdoses. The antidepressant study group. J
Emerg Med 1997; 15:439–445.
173. Robins MH. Survival following massive intoxication with Tofranil (imi-
pramine hydrochloride). J Am Osteopath Assoc 1971; 70:898–902.
174. Shannon M, Lovejoy FH, Jr. Pulmonary consequences of severe tricy-
clic antidepressant ingestion. J Toxicol Clin Toxicol 1987; 25:443–461.
175. Crome P, Dawling S, Braithwaite RA, Masters J, Walkey R. Effect of acti-
vated charcoal on absorption of nortriptyline. Lancet 1977; 2:1203–1205.
176. Bosse GM, Barefoot JA, Pfeifer MP, Rodgers GC. Comparison of
three methods of gut decontamination in tricyclic antidepressant over-
dose. J Emerg Med 1995; 13:203–209.
177. Hulten BA, Adams R, Askenasi R, Dallos V, Dawling S, Heath A,
Volans G. Activated charcoal in tricyclic antidepressant poisoning.
Hum Toxicol 1988; 7:307–310.
178. Hedges J, Otten E, Schroeder T, Tasset J. Correlation of initial
amitrpityline concentration reduction with activated-charcoal therapy
in overdose patients. Am J Emerg Med 1987; 5:48–51.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 229
179. Crome P, Adams R, Ali C, Dallos V, Dawling S. Activated charcoal in
tricyclic antidepressant poisoning: pilot controlled clinical trial. Hum
Toxicol 1983; 2:205–209.
180. Braithwaite RA, Crome P, Dawking S. The in vitro and in vivo evalua-
tion of activated charcoal as an adsorbent of tricyclic antidepressants.
[proceedings]. Br J Clin Pharmacol 1978; 7:368P.
181. Dawling S, Crome P, Braithwaite R. Effect of delayed administration
of activated charcoal on nortriptyline absorption. Eur J Clin Pharmacol
1978; 14:445–447.
182. Chyka PA, Seger D, Krenzelok EP, Vale JA; American Academy of
Clinical Toxicology; European Association of Poisons Centres and
Clinical Toxicologists. Position paper: Single-dose activated charcoal.
Clin Toxicol (Phila). 2005; 43:61–87.
183. Alaspaa AO, Kuisma MJ, Hoppu K, Neuvonen PJ. Out-of-hospital
administration of activated charcoal by emergency medical services.
Ann Emerg Med 2005; 45:207–212.
184. Lamminpaa A, Vilska J, Hoppu K. Medical charcoal for a child’s poi-
soning at home: availability and success of administration in Finland.
Hum Exp Toxicol 1993; 12:29–32.
185. Wax PM, Cobaugh DJ. Prehospital gastrointestinal decontamination
of toxic ingestions: a missed opportunity. Am J Emerg Med 1998;
16:114–116.
186. Godambe SA, Mack JW, Chung DS, Lindeman R, Lillehei CW, Colin
AA. Iatrogenic pleuropulmonary charcoal instillation in a teenager.
Pediatr Pulmonol 2003; 35:490–493.
187. Roy TM, Ossorio MA, Cipolla LM, Fields CL, Snider HL, Anderson
WH. Pulmonary complications after tricyclic antidepressant overdose.
Chest 1989; 96:852–856.
188. Ross JP, Small TR, Lepage PA. Imipramine overdose complicated by
toxic megacolon. Am Surg 1998; 64:242–244.
189. Wallace DE. Bowel ischemia in two patients following tricyclic anti-
depressant (TCA) overdose [abstract]. Vet Hum Toxicol 1989; 31:377.
190. Gomez HF, Brent JA, Munoz DCt, Mimmack RF, Ritvo J, Phillips S,
McKinney P. Charcoal stercolith with intestinal perforation in a patient
treated for amitriptyline ingestion. J Emerg Med 1994; 12:57–60.
191. Ray MJ, Radin DR, Condie JD, Halls JM, Padin DR. Charcoal bezoar.
Small-bowel obstruction secondary to amitriptyline overdose therapy.
Dig Dis Sci 1988; 33:106–107.
192. Knudsen K, Abrahamsson J. Epinephrine and sodium bicarbonate
independently and additively increase survival in experimental ami-
triptyline poisoning. Crit Care Med 1997; 25:669–674.
193. Albertson TE, Dawson A, de Latorre F, Hoffman RS, Hollander JE,
Jaeger A, Kerns WR, 2nd, Martin TG, Ross MP. TOX-ACLS: Toxico-
logic-oriented advanced cardiac life support. Ann Emerg Med 2001;
37:S78–90.
194. Brown TC. Sodium bicarbonate treatment for tricyclic antidepressant
arrhythmias in children. Med J Aust 1976; 2:380–382.
195. Hoffman JR, Votey SR, Bayer M, Silver L. Effect of hypertonic
sodium bicarbonate in the treatment of moderate-to-severe cyclic anti-
depressant overdose. Am J Emerg Med 1993; 11:336–341.
196. Bryan CK, Ludy JA, Hak SH, Roberts R, Marshall WR. Overdoses
with tricyclic antidepressants--two case reports. Drug Intell Clin
Pharm 1976; 10:380–384.
197. Citak A, Soysal DD, Ucsel R, Karabocuoglu M, Uzel N. Efficacy of
long duration resuscitation and magnesium sulphate treatment in ami-
triptyline poisoning. Eur J Emerg Med 2002; 9:63–66.
198. Fouron J, Chicoine R. ECG changes in fatal imipramine (Tofranil)
intoxication. Pediatrics 1971; 48:777–781.
199. Givens T, Holloway M, Wason S. Pulmonary aspiration of activated
charcoal: a complication of its misuse in overdose management. Pedi-
atr Emerg Care 1992; 8:137–140.
200. Glauser J. Tricyclic antidepressant poisoning. Cleve Clin J Med 2000;
67:704–706,709–713,717–709.
201. Greenblatt DJ, Koch-Weser J, Shader RI. Multiple complications and
death following protriptyline overdose. JAMA 1974; 229:556–557.
202. Guharoy SR. Adult respiratory distress syndrome associated with ami-
triptyline overdose. Vet Hum Toxicol 1994; 36:316–317.
203. Hodes D. Sodium bicarbonate and hyperventilation in treating an
infant with severe overdose of tricyclic antidepressant. Br Med J (Clin
Res Ed) 1984; 288:1800–1801.
204. Kingston ME. Hyperventilation in tricyclic antidepressant poisoning.
Crit Care Med 1979; 7:550–551.
205. Lin MH, Hung KL, Wang NK, Shen CT. Cardiotoxicity in imipramine
intoxication: report of one case. Acta Paediatr Taiwan 2001; 42:355–358.
206. Manoguerra AS. Tricyclic antidepressants. Crit Care Q 1982; 4:43–51.
207. Mehta NJ, Alexandrou NA. Tricyclic antidepressant overdose and
electrocardiographic changes. J Emerg Med 2000; 18:463–464.
208. Newton EH, Shih RD, Hoffman RS. Cyclic antidepressant overdose: a
review of current management strategies. Am J Emerg Med 1994;
12:376–379.
209. Orr DA, Bramble MG. Tricyclic antidepressant poisoning and pro-
longed external cardiac massage during asystole. Br Med J (Clin Res
Ed) 1981; 283:1107–1108.
210. Perel A, Cotev S. Imipramine (Tofranil) intoxication: a case report and
review of management. Crit Care Med 1976; 4:274–276.
211. Ramsay ID. Survival after imipramine poisoning. Lancet 1967;
2:1308–1309.
212. Sandeman DJ, Alahakoon TI, Bentley SC. Tricyclic poisoning--suc-
cessful management of ventricular fibrillation following massive over-
dose of imipramine. Anaesth Intensive Care 1997; 25:542–545.
213. Singh N, Singh HK, Khan IA. Serial electrocardiographic changes as a
predictor of cardiovascular toxicity in acute tricyclic antidepressant
overdose. Am J Ther 2002; 9:75–79.
214. Treitman P. Desipramine poisoning. JAMA 1972; 220:861,864.
215. Hoegholm A, Clementsen P. Hypertonic sodium chloride in severe
antidepressant overdosage. J Toxicol Clin Toxicol 1991; 29:297–298.
216. Lavoie FW, Gansert GG, Weiss RE. Value of initial ECG findings and
plasma drug levels in cyclic antidepressant overdose. Ann Emerg Med
1990; 19:696–700.
217. Liebelt EL, Ulrich A, Francis PD, Woolf A. Serial electrocardiogram
changes in acute tricyclic antidepressant overdoses. Crit Care Med
1997; 25:1721–1726.
218. Peters RW, Buser GA, Kim HJ, Gold MR. Tricyclic overdose causing
sustained monomorphic ventricular tachycardia. Am J Cardiol 1992;
70:1226–1228.
219. Rinder HM, Murphy JW, Higgins GL. Impact of unusual gastrointesti-
nal problems on the treatment of tricyclic antidepressant overdose.
Ann Emerg Med 1988; 17:1079–1081.
220. Shannon MW. Duration of QRS disturbances after severe tricyclic
antidepressant intoxication. J Toxicol Clin Toxicol 1992; 30:377–386.
221. Southall DP, Kilpatrick SM. Imipramine poisoning: survival of a child
after prolonged cardiac massage. Br Med J 1974; 4:508.
222. Teba L, Schiebel F, Dedhia HV, Lazzell VA. Beneficial effect of nore-
pinephrine in the treatment of circulatory shock caused by tricyclic
antidepressant overdose. Am J Emerg Med 1988; 6:566–568.
223. Tran TP, Panacek EA, Rhee KJ, Foulke GE. Response to dopamine vs.
norepinephrine in tricyclic antidepressant-induced hypotension. Acad
Emerg Med 1997; 4:864–868.
224. Rubenstein JS, Burg FD. Tricyclic antidepressant poisoning. Drug
Therapy 1989; 19:126–128,130.
225. Langou RA, Van Dyke C, Tahan SR, Cohen LS. Cardiovascular manifesta-
tions of tricyclic antidepressant overdose. Am Heart J 1980; 100:458–464.
226. McDuffee AT, Tobias JD. Seizure after flumazenil administration in a
pediatric patient. Pediatr Emerg Care 1995; 11:186–187.
227. Guidelines 2000 for Cardiopulmonary Resuscitation and Emergency
Cardiovascular Care. The American Heart Association in collabora-
tion with the International Liaison Committee on Resuscitation. Circu-
lation 2000; 102(suppl 8):1–370.
228. Sunshine P, Yaffe SJ. Amitriptyline poisoning. Clinical and pathologi-
cal findings in a fatal case. Am J Dis Child 1963; 106:501–506.
229. Halle MA, Collipp PJ. Amitriptyline hydrochloride poisoning. Unsuccess-
ful treatment by peritoneal dialysis. N Y State J Med 1969; 69:1434–1436.
230. Rollin P, Antaki AJ. Poisoning due to tricyclic antidepressants in chil-
dren and adolescents. Can Med Assoc J 1979; 120:951–956.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
230 A.D. Woolf et al.
231. Fendrick GM. Amitriptyline poisoning. N Engl J Med 1962;
267:1031–1032.
232. Brown TC, Barker GA, Dunlop ME, Loughnan PM. The use of
sodium bicarbonate in the treatment of tricyclic antidepressant-
induced arrhythmias. Anaesth Intensive Care 1973; 1:203–210.
233. Chambers T, Kindley AD. Amitriptyline poisoning in childhood. Br
Med J 1974; 3:687.
234. Noack CH. A death from overdosage of "Tofranil." Med J Aust 1960;
47(2):182.
235. Wright SP. Usefulness of physostigmine in imipramine poisoning. A
dramatic response in a child resistant to other therapy. Clin Pediatr
(Phila) 1976; 15:1123–1128.
236. Young GC, Morgan RT. Rapidity of response to the treatment of
enuresis. Dev Med Child Neurol 1973; 15:488–496.
237. Arneson GA. A near fatal case of imipramine overdosage. Am J Psy-
chiatry 1961; 117:934–936.
238. Ryan R, 3rd, Wians FH, Jr., Stigelman WH, Jr., Clark H, McCurdy F.
Imipramine poisoning in a child: lack of efficacy of resin hemoperfu-
sion. Pediatr Emerg Care 1985; 1:201–204.
239. Sueblinvong V, Wilson JF. Myocardial damage due to imipramine
intoxication. J Pediatr 1969; 74:475–478.
240. Jacobzinger H, Raybin HW. Imipramine hydrochloride intoxication. N
Y State J Med 1963; 63:1394–1398.
241. Fatteh A, Blanke R, Mann GT. Death from imipramine poisoning. J
Forensic Sci 1968; 13:124–128.
242. Parkin JM, Fraser MS. Poisoning as a complication of enuresis. Dev
Med Child Neurol 1972; 14:727–730.
243. Dingell JV, Sulser F, Gillette JR. Species differences in the metabo-
lism of Imipramine and desmethylimipramine (DMI). J Pharmacol
Exp Ther 1964; 143:14–22.
244. Thiemann HH, Otto L, Fritz H. Tödliche vergiftung durch imipramin.
Dtsch Gesundheitsw 1967; 22:1719–1722.
245. Prout BJ, Young J, Goddard P. Imipramine poisoning in childhood
and suggested treatment. Br Med J 1965; 5440:972.
246. Penny R. Imipramine hydrochloride poisoning in childhood. Am J Dis
Child 1968; 116:181–186.
247. Garrison HF, Jr., Moffitt EM. Imipramine hydrochloride intoxication.
JAMA 1962; 179:456–458.
248. Brown KG, McMichen HU, Briggs DS. Tachyarrhythmia in severe
imipramine overdose controlled by practolol. Arch Dis Child 1972;
47:104–106.
249. Sacks MH, Bonforte RJ, Laser RP, Dimich I. Cardiovascular compli-
cations of imipramine intoxication. JAMA 1968; 205:588–590.
250. Côté M, Elias G. Le propranolol dans les arythmes cardiaques par
intoxication à l’imipramine (Tofranil) chez l’enfant. Union Med Can
1974; 103:1223–1225.
251. Burks JS, Walker JE, Rumack BH, Ott JE. Tricyclic antidepressant
poisoning. Reversal of coma, choreoathetosis, and myoclonus by phy-
sostigmine. JAMA 1974; 230:1405–1407.
252. Sesso AM, Snyder RC, Schott CE. Propranolol in imipramine poison-
ing. Am J Dis Child 1973; 126:847–849.
253. Alajem N, Albagli C. Severe imipramine poisoning in an infant. Am J
Dis Child 1962; 103:702–705.
254. Braun L, Brodehl J, Fichsel H, Kallfelz C. Uber mipraminintoxikation
im kindesalter. Med Klin 1965; 60:1737–1742.
255. Connelly JF, Venables AW. A case of poisoning with “Tofranil”. Med
J Aust 1961; 1:109.
256. Oliver JS, Smith H. A case of fatal imipramine poisoning in an infant.
Med Sci Law 1977; 17:193–194.
257. Bickel MH, Brochon R, Friolet B, Herrmann B, Stofer AR. Clinical
and biochemical results of a fatal case of desipramine intoxication.
Psychopharmacologia 1967; 10:431–436.
258. Yang KL, Dantzker DR. Reversible brain death. A manifestation of
amitriptyline overdose. Chest 1991; 99:1037–1038.
259. Lindstrom FD, Flodmark O, Gustafsson B. Respiratory distress syn-
drome and thrombotic, non-bacterial endocarditis after amitriptyline
overdose. Acta Med Scand 1977; 202:203–212.
260. Bessen HA, Niemann JT. Improvement of cardiac conduction after
hyperventilation in tricyclic antidepressant overdose. J Toxicol Clin
Toxicol 1985; 23:537–546.
261. Manoguerra AS, Steiner RW. Prolonged neuromuscular blockade after
administration of physostigmine and succinylcholine. Clin Toxicol
1981; 18:803–805.
262. Gard H, Knapp D, Hanenson I, Walle T, Gaffney T. Studies on the dis-
position of amitriptyline and other tricyclic antidepressant drugs in
man as it relates to the management of the overdosed patient. Adv Bio-
chem Psychopharmacol 1973; 7:95–105.
263. Bolognesi R, Tsialtas D, Vasini P, Conti M, Manca C. Abnormal ven-
tricular repolarization mimicking myocardial infarction after heterocy-
clic antidepressant overdose. Am J Cardiol 1997; 79:242–245.
264. Flomenbaum N, Price D. Recognition and management of antidepressant
overdoses: tricyclics and trazodone. Neuropsychobiology 1986; 15:46–51.
265. Czech K, Francesconi M, Haimböck E, Hruby K. Die akute vergiftung
durch trizyklische antidepressiva und ihre therapie mit physostigmin-
salizylat. Wien Klin Wochenschr 1977; 89:265–269.
266. Borden EC, Rostand SG. Recovery from massive amitriptyline over-
dosage. Lancet 1968; 1:1256.
267. Oreopoulos DG, Lal S. Recovery from massive amitriptyline overdos-
age. Lancet 1968; 2:221.
268. LeWitt PA, Forno LS. Peripheral neuropathy following amitriptyline
overdose. Muscle Nerve 1985; 8:723–724.
269. Forbes G, Pollock Weir W, Smith H, Bogan J. Amitriptyline poison-
ing. J Forensic Sci Soc 1965; 5:183–187.
270. Hong WK, Mauer P, Hochman R, Caslowitz JG, Paraskos JA. Ami-
triptyline cardiotoxicity. Chest 1974; 66:304–306.
271. Ward FG, Tin-Myint B. Amitriptyline poisoning. Lancet 1965; 2:910.
272. Dequin PF, Lanotte R, Furet Y, Legras A, Perrotin D. Association bicar-
bonate molaire et adrénaline au cours d’une intoxication tricyclique
grave chez une femme gastrectomisée. Presse Med 1994; 23:540–541.
273. Stark JE, Bethune DW. Amitriptyline poisoning. Lancet 1965; 2:390.
274. Sunshine I, Baeumler J. A fatal case of poisoning with amitriptyline.
Nature 1963; 199:1103–1104.
275. Prenzel E, Krohs G. Intoxikationen mit trizyklischen antidepressiva
und phenothiazinen sowie deren therapie mit physostigmin. Anaesthe-
siol Reanim 1986; 11:227–234.
276. Walsh DM. Cyclic antidepressant overdose in children: a proposed
treatment protocol. Pediatr Emerg Care 1986; 2:28–35.
277. Lloyd TW, Hart DR. Amitriptyline poisoning. Lancet 1965; 2:544.
278. Jeong YG, Caccamo LP. Amitriptyline poisoning causing left bundle
branch block. Ohio State Med J 1976; 72:217–219.
279. Mehrotra TN. Amitriptyline poisoning. Lancet 1965; 2:544–545.
280. Davies DM, Allaye R. Amitriptyline poisoning. Lancet 1963; 2:543.
281. Leys D, Pasquier F, Lamblin MD, Dubois F, Petit H. Acute polyradiculoneur-
opathy after amitriptyline overdose. Br Med J (Clin Res Ed) 1987; 294:608.
282. Ordiway MV. Treating tricyclic overdose with physostigmine. Am J
Psychiatry 1978; 135:1114.
283. Hurst HE, Jarboe CH. Clinical findings, elimination pharmacokinetics,
and tissue drug concentrations following a fatal amitriptyline intoxica-
tion. Clin Toxicol 1981; 18:119–125.
284. Babb SV, Dunlop SR. Case report of sudden and unexpected death
after tricyclic overdose. Am J Psychiatry 1985; 142:275–276.
285. Roberge RJ, Krenzelok EP. Prolonged coma and loss of brainstem reflexes
following amitriptyline overdose. Vet Hum Toxicol 2001; 43:42–44.
286. Knudsen K, Abrahamsson J. Magnesium sulphate in the treatment of ventric-
ular fibrillation in amitriptyline poisoning. Eur Heart J 1997; 18:881–882.
287. Nicholls HK. Amitriptyline overdose and the ECG: report of a case.
NZ Med J 1965; 64:651–652.
288. Holinger PC, Klawans HL. Reversal of tricyclic-overdosage-induced
central anticholinergic syndrome by physostigmine. Am J Psychiatry
1976; 133:1018–1023.
289. Bain DJ, Turner T. Imipramine poisoning. Arch Dis Child 1971; 46:887.
290. Bailey RR, Sharman JR, O’Rourke J, Buttimore AL. Haemodialysis
and forced diuresis for tricyclic antidepressant poisoning. Br Med J
1974; 4:230–231.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 231
291. Johnson DA, Knepp IG, Whelan TV. Toxic tricyclic antidepressant
levels and the ECG. JAMA 1983; 250:1027.
292. Harthorne JW, Marcus AM, Kaye M. Management of massive imi-
pramine overdosage with mannitol and artificial dialysis. N Engl J
Med 1963; 268:33–36.
293. Rasmussen J. Amitriptyline and imipramine poisoning [letter]. Lancet
1965; 2:850–851.
294. Rushnak MJ, McGovern DP. Reversal of imipramine cardiotoxicity
with physostigmine. J Med Soc N J 1977; 74:155–157.
295. Reed K, McKim HR. ECG changes in pure impramine overdose as
function of plasma level. Can Psychiatr Assoc J 1978; 23:573–577.
296. Freimuth HC. Poisoning by new drugs – report of a fatality due to sui-
cidal ingestion of tofranil. J Forensic Sci 1961; 6: 68–75.
297. Lancaster NP, Foster AR. Suicidal attempt by imipramine overdosage.
Br Med J 1959; 5164:1458.
298. Dubuc M, Friborg J, Houde M, Laplante L. Traitement de l’intoxica-
tion médicamenteuse aux agents antidépresseurs tricycliques: pour ou
contre l’utilisation de la physostigmine. Union Med Can 1981;
110:555–557.
299. Dolara P, Franconi F. Hypertonic sodium chloride and lidocaine in a
case of imipramine intoxication. Clin Toxicol 1977; 10:395–398.
300. Herson VC, Schmitt BD, Rumack BH. Magical thinking and imi-
pramine poisoning in two school-age children. JAMA 1979;
241:1926–1927.
301. Denton S. “Tofranil” (imipramine) in toxicological analysis. Analyst
1962; 87:234–236.
302. Edwards J. Fatal imipramine overdosage. Med J Aust 1966; 1:839–840.
303. Louis C, Olbing H, Bohlmann HG, Philippou A, Heimsoth V. Zur
behandlung der imipramin-vergiftung beim kind. Dtsch Med Wochen-
schr 1970; 95:2078–2082.
304. Lee FI. Imipramine overdosage--report of a fatal case. Br Med J 1961;
5222:338–339.
305. Hoffman JR, McElroy CR. Bicarbonate therapy for dysrhythmia and
hypotension in tricyclic antidepressant overdose. West J Med 1981;
134:60–64.
306. Pearson JD, Jones ES, Gabbe DM. Cardiac arrest and arrhythmias due to
self-poisoning with imipramine. Anaesthesia 1969; 24:69–71.
307. Bismuth C, Pontal PG, Baud F, Galliot M, Elkhouly M. Prolonged
high plasma imipramine levels after acute intoxication. Vet Hum Tox-
icol 1982; 24:69–70.
308. Kirchmair H, Goldberg K. Suicidver such mit Tofranil in kombination
mit schlafmitteln. Med Libre 1960; 55:1474–1475.
309. Bindler RM, Howry LB. Prentice Hall Pediatric Drug Guide. Upper
Saddle River, NJ: Pearson/Prentice Hall, 2005.
310. Physicians’ Desk Reference: PDR. 60th ed. Montvale, NJ: Thomson
Healthcare, 2006.
311. Takemoto CK, Hodding JH, Kraus DM. Pediatric Dosage Handbook.
12th ed. Hudson, OH: Lexi-Comp, 2005.
Appendix 1
Expert consensus panel members
Lisa L. Booze, Pharm.D.
Certified Specialist in Poison Information
Maryland Poison Center
University of Maryland School of Pharmacy
Baltimore, Maryland, USA
E. Martin Caravati, M.D., M.P.H., FACMT, FACEP
Professor of Surgery (Emergency Medicine)
University of Utah
Medical Director
Utah Poison Center
Salt Lake City, Utah, USA
Gwenn Christianson, R.N., M.S.N.
Certified Specialist in Poison Information
Indiana Poison Center
Indianapolis, Indiana, USA
Peter A. Chyka, Pharm.D., FAACT, DABAT
Professor, Department of Clinical Pharmacy
College of Pharmacy
University of Tennessee Health Science Center
Memphis, Tennessee, USA
Daniel J. Cobaugh, Pharm.D., FAACT, DABAT
Director of Research
American Society of Health-System Pharmacists Research
and Education Foundation
Bethesda, Maryland, USA
Former Associate Director, American Association of Poison
Control Centers
Anthony S. Manoguerra, Pharm.D., DABAT, FAACT
Professor of Clinical Pharmacy and Associate Dean
School of Pharmacy and Pharmaceutical Sciences
University of California San Diego
Former Director, California Poison Control System, San Diego
Division
San Diego, California, USA
Lewis S. Nelson, M.D., FACEP, FACMT
Associate Professor of Emergency Medicine
New York University School of Medicine
Associate Medical Director
New York City Poison Control Center
New York, New York, USA
Kent R. Olson, M.D., FACEP, FAACT, FACMT
Medical Director
California Poison Control System, San Francisco Division
Clinical Professor of Medicine & Pharmacy
University of California, San Francisco
San Francisco, California, USA
Elizabeth J. Scharman, Pharm.D., DABAT, BCPS, FAACT
Director, West Virginia Poison Center
Professor, West Virginia University School of Pharmacy
Department of Clinical Pharmacy
Charleston, West Virginia, USA
Paul M. Wax, M.D., FACMT
Attending Toxicologist
UT Southwestern Medical Center
Dallas, Texas, USA
Alan D. Woolf, M.D., M.P.H., FACMT
Director, Program in Environmental Medicine
Children’s Hospital, Boston
Associate Professor of Pediatrics
Harvard Medical School
Boston, Massachusetts, USA
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
232 A.D. Woolf et al.
Appendix 2
Grades of recommendation and levels of evidence
Appendix 3
Secondary review panel organizations
Ambulatory Pediatric Association
American Academy of Breastfeeding Medicine
American Academy of Emergency Medicine
American Academy of Pediatrics
American Association for Health Education
American College of Clinical Pharmacy
American College of Emergency Physicians
American College of Occupational and Environmental
Medicine
American Pharmacists Association
American Public Health Association
American Society of Health-System Pharmacists
Association of Maternal and Child Health Programs
Association of Occupational and Environmental Clinics
Association of State and Territorial Health Officials
Canadian Association of Poison Control Centres
Centers for Disease Control and Prevention – National
Center for Injury Prevention and Control
Consumer Federation of America
Consumer Product Safety Commission
Department of Transportation
Emergency Medical Services for Children
Emergency Nurses Association
Environmental Protection Agency
Food and Drug Administration
National Association of Children’s Hospitals and
Related Institutions
National Association of Emergency Medical Services
Physicians
National Association of Emergency Medical
Technicians
National Association of School Nurses
National Association of State Emergency
Medical Services Directors
National Safe Kids Campaign
Teratology Society
World Health Organization International Programme on
Chemical Safety
Grade of
recommendation Level of evidence Description of study design
A 1a Systematic review (with homogeneity) of randomized clinical trials
1b Individual randomized clinical trials (with narrow confidence interval)
1c All or none (all patients died before the drug became available,
but some now survive on it; or when some patients died before
the drug became available, but none now die on it.)
B 2a Systematic review (with homogeneity) of cohort studies
2b Individual cohort study (including low quality randomized clinical trial)
2c “Outcomes” research
3a Systemic review (with homogeneity) of case-control studies
3b Individual case-control study
C 4 Case series, single case reports (and poor quality cohort and case control studies)
D 5 Expert opinion without explicit critical appraisal or based on
physiology or bench research
Z 6 Abstracts
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
Tricyclic antidepressant poisoning 233
Appendix 4
Algorithm for triage of tricyclic antidepressant ingestion
Is suicidal, abuse, or malicious intent suspected?
YES Refer to emergency department.
NO
Is the home situation of concern? (e.g., patient lives alone or family/caregiver
seems unreliable)
YES Refer to emergency department.
NO
Is the patient symptomatic? (e.g., weak, drowsy, dizzy, tremulous,
palpitations)
YES Refer to emergency department.
NO
Have more than 6 hours elapsed since the TCA ingestion and the patient is
still asymptomatic?
YES Continue to follow closely at home.
NO
Does the patient have significant underlying cardiovascular or neurological
disease, or is he/she taking a cardiodepressant drug or MAO inhibitor?
YES Consider referral to emergency department.
NO
Can you estimate the maximum amount ingested?
NO Refer to emergency department.
YES
Has the patient ingested a potentially toxic dose?
*
YES Refer to emergency department.
Amitriptyline >5 mg/kg
Clomipramine >5 mg/kg
Desipramine >2.5 mg/kg
Doxepin >5 mg/kg
Doxepin cream
>5 mg/kg
Imipramine >5 mg/kg
Nortriptyline >2.5 mg/kg
Protriptyline >1 mg/kg
Trimipramine >2.5 mg/kg
NO
Observe at home. Instruct caller to call poison center back if symptoms
appear. Consider poison center-initiated follow-up within 4 hours of initial
call. Consider referral to emergency services should new symptoms develop.
*Algorithm applies only to ingested TCAs, not to parenteral use or other routes of exposure. Algorithm applies only to acute ingestions.
A toxic dose for dermal exposures could not be established from available evidence. The dose represents the ingestion of a dermal preparation.
Clinical Toxicology Downloaded from informahealthcare.com by 117.174.223.37 on 05/20/14
For personal use only.
... Emergency department statistics indi-cate that tricyclic antidepressants (TCAs) drugs are frequently misused for suicidal purposes (1). TCAs stand out as one of the leading causes of drug poisoning (2), despite the availability of safer alternatives with a less toxic profile, 2 such as selective serotonin reuptake inhibitors (SSRIs). Surprisingly, TCAs continue to be prescribed extensively for various indications in both children and adults, (3) ranging from major depressive disorders to chronic pain syndromes, panic attacks, severe anxiety, phobic disorders, obsessive-compulsive disorders, migraine, eating disorders, and attention deficit hyperactivity disorders (4). ...
... Surprisingly, TCAs continue to be prescribed extensively for various indications in both children and adults, (3) ranging from major depressive disorders to chronic pain syndromes, panic attacks, severe anxiety, phobic disorders, obsessive-compulsive disorders, migraine, eating disorders, and attention deficit hyperactivity disorders (4). Their mechanism involves primarily blocking norepinephrine and postsynaptic α-adrenergic receptors while also influencing the cholinergic and histaminergic systems and α-adrenergic activity, along with quinidine-like effects on cardiac condition (2,3,5). Tricyclic antidepressants possess a narrow therapeutic index, primarily responsible for inducing adverse effects and accounting for a significant proportion of severe poisoning cases after TCAs overdose. ...
... Tricyclic antidepressants possess a narrow therapeutic index, primarily responsible for inducing adverse effects and accounting for a significant proportion of severe poisoning cases after TCAs overdose. The continued use of TCA necessitates careful consideration of their potential toxicity and the need for effective prevention strategies (2,5). In the early stages of poisoning, symptoms such as dry mouth, blurred vision, sinus tachycardia, and drowsiness arise due to anticholinergic activity, while drowsiness results from antihistaminergic activity (6). ...
... However, TCA may cause cardiotoxicity that leads to high rates of mortality and morbidity, and AMT is the most common agent causing TCA toxicity. 5,6 AMT may cause cardiotoxicity due to ventricular arrhythmias caused by its Na channel inhibition and changes in intracellular Ca metabolism. 7,8 The toxicity caused by TCA is dose independent and this toxicity manifests itself with prolongation in PR, QT and QTc intervals, measured on the ECG. 9 QT prolongation on ECG is a predictor for toxicity and indicates a poor prognosis. ...
... In addition to the wide clinical uses of TCA, the cardiotoxicity caused by the use of these drugs limits the use of all TCAs, primarily AMT. 6 AMT may cause cardiotoxicity due to ventricular arrhythmias caused by the prolongation of the QRS, QTc and PR segments, as seen on ECG, as a result of Na channel inhibition caused by AMT. 7 Since the cardiotoxicity caused by AMT is not dose dependent, it is important to monitor the ECG findings. 23 In our study, we found that EMPA significantly prevented AMT-induced QTc prolongation. ...
Article
Aim: Empagliflozin (EMPA) is a sodium-glucose transporter-2 inhibitor used in the treatment of type 2 diabetes and has positive effects on cardiovascular outcomes. Amitriptyline (AMT) can be used in many clinical indications but leads to cardiotoxicity by causing QT prolongation. Our aim in this study was to determine how the effects of the concomitant use of empagliflozin and amitriptyline, which have been shown to have effects on sodium and calcium metabolism in cardiomyocytes, would cause an effect on QT and QTc intervals in clinical practice. Methods: Twenty-four male Wistar albino rats were randomised into four groups. The control group received only physiological serum (1 ml) via orogastric gavage (OG). The EMPA group received empagliflozin (10 mg/kg) via OG. The AMT group received amitriptyline (100 mg/kg) via OG. The AMT + EMPA group (n = 6) received amitriptyline (100 mg/kg) and empagliflozin (10 mg/kg). Under anaesthesia, QT and QTc intervals were measured at baseline, and in the first and second hours. Results: In the AMT group, QT intervals and QTc values were found to be statistically longer than in the control group (p ≤ 0.001). Empagliflozin significantly ameliorated amitriptyline-induced QT and QTc prolongation. In the AMT + EMPA group, QT and QTc intervals were significantly lower compared to that in the AMT group (p < 0.01). Conclusion: In this study, we determined that empagliflozin significantly ameliorated amitriptyline-induced QT and QTc prolongation. This effect was probably due to the opposite effects of these two agents in the intracellular calcium balance. With more clinical trials, the routine use of empagliflozin may be suggested to prevent QT and QTc prolongation in diabetic patients receiving amitriptyline.
... Toxins in biology are substances that cause disturbances to live organisms, and this often occurs either through chemical reactions or others at the molecular level in the body. Natural toxins can be produced through the accumulation of some chemical substances and elements that the animal gets from food [1], they come as residues from dilapidated cells or some vital processes in the body, causing sheep inside the body that acts as a substance that cannot be used in the metabolic processes and difficult to get rid of [2]. [3] indicated that the animal is able to get rid of these substances accumulated inside the body that are difficult to get rid of in the usual ways through a mechanism called Glucuronide, known as Glucuronoside [4]. ...
Article
Full-text available
The sheep like other animals need more and more welfare to produce meat of good quality, Sheep welfare problems primarily arise from mutilations, lameness, transportation, and disease. With proper grazing, breeding, and husbandry practices, most health issues in sheep can be resolved or avoided. Grazing involves seeking and choosing feed as well as grabbing and consuming it. The manner in which ruminants or grazing animals grip and consume forages varies. These variations are connected to the sorts of fodder that various ruminants prefer. Heat stress, cold stress, weariness, prolonged thirst, prolonged hunger, hampered movement, movement restrictions, resting issues, social stress, pain, fear, and distress are the welfare types that sheep may encounter during caged, all issues due to shortage of welfare, this results in sheep inside the body that acts as a substance that cannot be used in the metabolic processes and is difficult to get rid of the animal is able to get rid of these substances accumulated inside the body that are difficult to get rid of the presence of natural toxins can be produced through the accumulation of some chemical substances and elements that the animal gets from the food. They come as residues from deteriorated cells or some vital processes in the body.
... Some adolescents may play such games because their anger levels are high. Result of a study on violent games have shown that children may be at higher risk [46]. It is known that many children use games for emotional regulation to relax, forget problems or feel less alone [47]. ...
Article
Full-text available
Objective: In this study, it was aimed to determine the relationship between the causes and duration of gaming and types of games and levels of anger among Turkish adolescents studying at high schools. Materials and Methods: The study was conducted using a cross-sectional descriptive design with adolescents enrolled in high schools in an Eastern Turkish province. A total of 819 adolescent students aged 13-18 were included in the study. Data were collected online through Google Forms, utilizing both the “Sociodemographic Form” and the “Adolescent Anger Rating Scale.” Results: The mean age of the adolescents was 15.52±1.29. Gender, academic performance, daily sleep duration, and daily walking distance were not found to be associated with anger levels. Additionally, there was no significant correlation between anger scores and the status of computer and smartphone usage. However, individuals who used smartphones for 3 hours or more for purposes such as gaming, entertainment, chatting, messaging, and socializing exhibited higher anger scores. Conclusion: Academic grades, the duration of smartphone usage, computer and smartphone use for gaming/entertainment, chatting, messaging, and socializing were found to be associated with anger. Specifically, extended periods of playing war, fighting, and similar games on both computers and smartphones were correlated with higher levels of anger. It would be advantageous for parents to monitor and regulate the content of the games their adolescents play, observe any behavioral differences, and take necessary precautions.
... Tricyclic antidepressants have a relatively low therapeutic index, as the toxic effects appear at drug concentrations only 3 times higher than the maximal prescribed dose. 12,13 For this and other reasons, the medications of this class, in particular amitriptyline, are notoriously known as one of the most common agents of an intentional drug overdose, resulting in 395 fatal cases in England and Wales over 2000−2006 14 and 784 fatal cases over 2007−2014, with a fatality risk close to 5%. 15 The use of sustained-release formulations delays the time to reach toxic concentration and, therefore, prolongs the period for the first aid to be effective. ...
... Antidepressants are commonly used to treat various types of depression and mental illnesses. Many important accidents are due to disorders related to these antidepressants [2]. On the other hand, excessive consumption of these antidepressants causes side effects such as diarrhea, nausea, and hypertension, and sometimes even leads to human death. ...
Article
Full-text available
Composite nanofibers, namely, polyvinyl alcohol (PVA), citric acid (CA), β-cyclodextrin (β–CD), and copper oxide nanoparticles (PVA/CA/β-cyclodextrin/CuO NPs), were developed as a novel, green, and efficient adsorbent in the pipette tip-micro-solid-phase extraction method (PT-µSPE), for the simultaneous extraction of three antidepressants drugs namely imipramine (IMP), citalopram (CIT), and clozapine (CLZ) in biological fluids before quantification by gas chromatography (GC-FID). Based on the obtained results from field emission scanning electron microscopy (FE-SEM), energy-dispersive X-ray spectroscopy (EDX), Fourier transform infrared spectroscopy (FT-IR), and X-ray diffraction (XRD), the successful synthesis of composite nanofibers was approved. Due to the presence of β-cyclodextrins and CuO NPs rich of functional groups on their surface, the nanofibers have high extraction efficiency. Under the optimal conditions, the linear range for imipramine, citalopram, and clozapine was 0.1 to 1000.0 ng mL⁻¹ with a determination coefficient ≥ 0.99. The limits of detection (LODs) were in the range 0.03 to 0.15 ng mL⁻¹. The relative standard deviation was 4.8 to 8.7% (within-day, n = 4) and 5.1 to 9.2% (between-day, n = 3) for 3 consecutive days. In addition, excellent clean-up was achieved which is a great advantage over other sample preparation methods. Finally, the ability of the developed method to extract the target analytes from the biological samples was evaluated. Graphical Abstract
Article
Full-text available
Flumazenil, a benzodiazepine antagonist, holds promise as a rapid intervention for reversing benzodiazepine overdose. However, its clinical utility is nuanced, characterized by complexities and potential risks. This review explores the multifaceted landscape of flumazenil use, encompassing its mechanisms of action, indications, adverse effects, monitoring protocols, toxicity, and outcomes. Additionally, it examines the interplay between flumazenil and benzodiazepine dependence, seizure risk, and co-ingestions. Through an interdisciplinary lens, we navigate the clinical considerations and challenges surrounding flumazenil therapy, providing insights to optimize patient care.
Article
Doxepin, dosulepin, and clomipramine are tricyclic antidepressants (TCAs) that act as serotonin and noradrenaline reuptake inhibitors. The metabolites formed by N-dealkylation of these tricyclic antidepressants contribute to overall poor pharmacokinetics and efficacy. Deuteration of the methyl groups at metabolically active sites has been reported to be a useful strategy for developing more selective and potent antidepressants. This isotopic deuteration can lead to better bioavailability and overall effectiveness. The objective is to study the effect of site-selective deuteration of TCAs on their pharmacokinetic and pharmacodynamic profile by comparison with their nondeuterated counterparts. In the current study, the pharmacokinetic profile and antidepressant behavior of deuterated TCAs were evaluated using the forced swim test (FST) and tail suspension test (TST), using male Wistar rats and male Swiss albino mice, respectively; additionally, a synaptosomal reuptake study was carried out. Compared with the nondeuterated parent drugs, deuterated forms showed improved efficacy in the behavior paradigm, indicating improved pharmacological activity. The pharmacokinetic parameters indicated increased maximum concentration in the plasma (Cmax), elimination half-life (t1/2), and area under the concentration-time curve (AUC) in deuterated compounds. This can have a positive clinical impact on antidepressant treatment. Synaptosomal reuptake studies indicated marked inhibition of the reuptake mechanism of serotonin (5-HT) and norepinephrine. Deuterated TCAs can prove to be potentially better molecules in the treatment of neuropsychiatric disorders as compared with nondeuterated compounds. In addition, we have demonstrated a concept that metabolically active, site-selective deuteration can be beneficial for improving the pharmacokinetic and pharmacodynamic profiles of TCAs. A further toxicological study of these compounds is needed to validate their future clinical use.
Article
Introduction: Chronic tricyclic antidepressant toxicity is rarely described in children. Symptoms include confusion, ataxia, and seizures. Toxicity may result from dosing error, CYP2C19 and CYP2D6 genetic variability, and drug-drug interactions. Chronic doxepin toxicity has not been previously reported in children. Doxepin is prescribed for insomnia and depression, with a maximum off-label dose of 3 mg/kg in children. We present a case of chronic doxepin toxicity mimicking epilepsy in a child attributable to three potential factors: supratherapeutic dosing, pharmacogenomic variability, and drug-drug interactions. Case report: A 10-year-old boy with insomnia, diagnosed with epilepsy 6 months prior, presented to an emergency department with confusion, ataxia, and increasing seizure frequency. He was prescribed doxepin for insomnia and four antiepileptics for seizures. After admission, he had two seizures and remained confused. EKGs showed QRS prolongation, suggesting doxepin toxicity. Doxepin-nordoxepin combined serum concentration was 1419 ng/mL (therapeutic 100-300 ng/mL), confirming doxepin toxicity. Outpatient records showed onset of confusion and seizures as doxepin dose was gradually uptitrated to 300 mg nightly (4.41 mg/kg). Symptoms worsened following addition of clobazam (CYP2D6 inhibitor) and topiramate (CYP2C19 inhibitor). Following doxepin discontinuation, all symptoms resolved. CYP2D6 testing showed intermediate metabolizer phenotype (CYP2D6*1/*4; activity score = 1.0; copy number = 2.0). No seizures have occurred in more than one year since doxepin discontinuation. Discussion: Caution must be exercised when prescribing doxepin. Pharmacogenomics, dose, drug-drug interactions, and age should be considered. Chronic toxicity should be contemplated in patients taking doxepin without acute overdose who present with persistent neurologic abnormalities including seizure.
Article
Clomipramine, a tricyclic antidepressant used to treat depression and obsessive-compulsive disorder, has been linked to a few cases of acute hepatotoxicity. It is also recognized as a compound that hinders the functioning of mitochondria. Hence, the effects of clomipramine on mitochondria should endanger processes that are somewhat connected to energy metabolism in the liver. For this reason, the primary aim of this study was to examine how the effects of clomipramine on mitochondrial functions manifest in the intact liver. For this purpose, we used the isolated perfused rat liver, but also isolated hepatocytes and isolated mitochondria as experimental systems. According to the findings, clomipramine harmed metabolic processes and the cellular structure of the liver, especially the membrane structure. The considerable decrease in oxygen consumption in perfused livers strongly suggested that the mechanism of clomipramine toxicity involves the disruption of mitochondrial functions. Coherently, it could be observed that clomipramine inhibited both gluconeogenesis and ureagenesis, two processes that rely on ATP production within the mitochondria. Half-maximal inhibitory concentrations for gluconeogenesis and ureagenesis ranged from 36.87μM to 59.64μM. The levels of ATP as well as the ATP/ADP and ATP/AMP ratios were reduced, but distinctly, between the livers of fasted and fed rats. The results obtained from experiments conducted on isolated hepatocytes and isolated mitochondria unambiguously confirmed previous propositions about the effects of clomipramine on mitochondrial functions. These findings revealed at least three distinct mechanisms of action, including uncoupling of oxidative phosphorylation, inhibition of the FoF1-ATP synthase complex, and inhibition of mitochondrial electron flow. The elevation in activity of cytosolic and mitochondrial enzymes detected in the effluent perfusate from perfused livers, coupled with the increase in aminotransferase release and trypan blue uptake observed in isolated hepatocytes, provided further evidence of the hepatotoxicity of clomipramine. It can be concluded that impaired mitochondrial bioenergetics and cellular damage are important factors underlying the hepatotoxicity of clomipramine and that taking excessive amounts of clomipramine can lead to several risks including decreased ATP production, severe hypoglycemia, and potentially fatal outcomes.
Article
The use of physostigmine in the treatment of tricyclic antidepressant overdose is discussed. The importance of continuous observation is emphasized and the method of preparation of physostigmine is outlined.
Article
The authors present a 46-year-old female and a 14-month-old female following ingestion of an overdose of tricyclic antidepressants, amitriptyline and imipramine, respectively. The discussion, following the cases, reviews the toxic manifestations of the antidepressive agents, i.e., CNS depression, agitation, delirium, coma, cardiac manifestations and anticholinergic effects. Review of appropriate treatment, particularly the use of physostigmine, is also presented.
Article