You are on page 1of 8

Food and Chemical Toxicology 142 (2020) 111393

Contents lists available at ScienceDirect

Food and Chemical Toxicology


journal homepage: www.elsevier.com/locate/foodchemtox

Review

Phenytoin – An anti-seizure drug: Overview of its chemistry, pharmacology T


and toxicology
Jiri Patockaa,b, Qinghua Wuc,d, Eugenie Nepovimovac, Kamil Kucab,c,∗
a
Faculty of Health and Social Studies, Department of Radiology and Toxicology, University of South Bohemia Ceske Budejovice, Ceske Budejovice, Czech Republic
b
Biomedical Research Centre, University Hospital, Hradec Kralove, Czech Republic
c
Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
d
College of Life Science, Yangtze University, Jingzhou, 434025, China

ARTICLE INFO ABSTRACT

Keywords: Phenytoin is a long-standing, anti-seizure drug widely used in clinical practice. It has also been evaluated in the
Phenytoin context of many other illnesses in addition to its original epilepsy indication. The narrow therapeutic index of
Pharmacokinetics phenytoin and its ubiquitous daily use pose a high risk of poisoning. This review article focuses on the chemistry,
Toxicity pharmacokinetics, and toxicology of phenytoin, with a special focus on its mutagenicity, carcinogenicity, and
Side effect
teratogenicity. The side effects on human health associated with phenytoin use are thoroughly described. In
DRESS syndrome
Cerebellar atrophy
particular, DRESS syndrome and cerebellar atrophy are addressed. This review will help in further under-
standing the benefits phenytoin use in the treatment of epilepsy.

1. Introduction of status epilepticus, especially where no further improvement is ob-


served with benzodiazepines (Prasad et al., 2014). Occasionally, neu-
Phenytoin is an anti-seizure drug that has been under clinical eva- ropathic pain or heart arrhythmias are also treated in this fashion
luation for around eight decades. It is primarily used for the treatment (Somberg, 1985; Kopsky et al., 2017). In addition, neurological, psy-
of tonic-clonic and partial seizures (Abou-Khalil, 2016). Phenytoin has chiatric, and non-CNS indications of phenytoin have been identified,
a highly selective inhibitory effect on the motor area of the cerebral such as wound healing, and several of these indications have been in-
cortex. For its mode of action, phenytoin binds to the inactivated state vestigated in subsequent pilot studies (Şimşek et al., 2014). Wound
of the Na+ channel to prolong the neuronal refractory period healing is one of the indications most thoroughly investigated (Keppel
(Hesselink, 2017). Moreover, it is generally believed that phenytoin Hesselink, 2018; Barratt et al., 1997).
exerts its antiepileptic effect by stabilising the function of brain cell However, phenytoin treatment is associated with several side ef-
membranes and increasing the levels of the inhibitory neuro- fects, and toxicity plays a key role in these issues (Imam et al., 2014).
transmitters serotonin (5-HT) and γ-aminobutyric acid (GABA) in the An excessive dose of phenytoin inhibits the central nervous system,
brain (Hesselink, 2017; Keppel Hesselink and Kopsky, 2017a,b). causes cerebellar dysfunction, dyskinesia, and induces peripheral neu-
Through these mechanisms, phenytoin prevents the spread of abnormal ropathy. In addition, phenytoin can cause gingival hyperplasia, induce
discharge and has anti-epileptic effects. The anti-neuralgia activity of pseudolymphomas and malignant lymphomas, and cause allergic re-
phenytoin may also be related to these same mechanisms; a reduction actions (Hesselink, 2017; Farook et al., 2019). As mentioned above,
in synaptic transmission or a reduction in transient stimuli that cause intravenous injections of phenytoin slow the conduction rate between
neuronal discharge (Hall et al., 2020). In cardiac tissue, phenytoin in- myocardial fibres and eliminate ectopic rhythms, producing a similar
hibits the ectopic rhythm of the atrium and ventricle, and accelerates effect to quinidine (Mathews et al., 2019). In addition, phenytoin can
the conduction of the atrioventricular node to reduce myocardial au- cause cerebellar syndrome, which manifests itself as ataxia, nystagmus,
tonomy, producing an antiarrhythmic effect (Zhang et al., 2018). hand tremor, and diplopia. These manifestations of cerebellar syn-
Phenytoin can be administered by oral delivery or parenteral de- drome are dependent on phenytoin dose and blood concentration;
livery. The intravenous version of this drug is usually made available as tremor occurs at an average plasma concentration of 20 μg/mL, and
a sodium salt, and this can be adapted to a propylene glycol-alcohol- ataxia occurs at 30 μg/mL. A coma can occurred at average plasma
water solution for parenteral administration of phenytoin for treatment concentrations of 40 μg/mL or more (Zhang et al., 2018). In the long-


Corresponding author. Biomedical Research Centre, University Hospital, Hradec Kralove, Czech Republic.
E-mail addresses: kamil.kuca@uhk.cz, kamil.kuca@fnhk.cz (K. Kuca).

https://doi.org/10.1016/j.fct.2020.111393
Received 1 January 2020; Received in revised form 16 April 2020; Accepted 24 April 2020
Available online 04 May 2020
0278-6915/ © 2020 Published by Elsevier Ltd.
J. Patocka, et al. Food and Chemical Toxicology 142 (2020) 111393

term, sudden seizures may become worse during phenytoin treatment, produced by oxidation of benzoin with nitric acid to produce the
although seizures can become more worse following withdrawal of this benzyl, after which the procedure is the same as for Biltz synthesis. In
medicine. this process, almonds may be used as a source of benzaldehyde
Phenytoin is more and more widely used in clinical practice, in- (Ashnagar et al., 2009). Water-soluble phenytoin derivatives have also
cluding for wound healing, migraine, dizziness, hiccups, myocardial been synthesised (Bosch et al., 1999). It should be noted that during
infarction, and burns (Keppel Hesselink and Kopsky, 2017a; b). decomposition caused by heat, phenytoin typically produces several
Therefore, the development and utilisation of this drug has attracted toxic fumes, including nitrogen oxide, carbon monoxide and carbon
increasing attention. However, the adverse reactions associated with dioxide (Lewis, 2004).
phenytoin treatment should not be ignored; known symptoms include
gastrointestinal irritation, gum hyperplasia, allergic reactions, and 4. Mechanism of action
mental health issues (Sadeghi et al., 2018). To prevent such serious
adverse consequences, it is necessary that phenytoin is used reasonably The mechanism of action of phenytoin in neuropharmacology has
in clinical practice. This review article focuses on the chemistry, me- been investigated for more than 80 years (Hesselink, 2017). Phenytoin
chanism of action, pharmacokinetics, toxicology of phenytoin, and its is a voltage-gated, sodium channel blocker (Hains et al., 2004). It exerts
side effects on human health. its effect by stabilising the inactive state of the Na+ channel and
prolonging the neuronal refractory period (Hesselink, 2017; Keppel
2. History Hesselink and Kopsky, 2017a; b). Through this mode of action, phe-
nytoin regulates the bioelectrical activities of various systems in hu-
The German chemist Heinrich Biltz synthesised phenytoin (diphe- mans. Importantly, phenytoin inhibits or eliminates abnormal electrical
nylhydantoin) for the very first time in 1908. He later sold his work to activity in nerve and muscle cells without affecting the production and
Parke-Davis (Keppel Hesselink and Kopsky, 2017a,b). The additional conduction of normal bioelectricity. The electrophysiological basis of
usefulness of phenytoin was revealed in 1938 by Tracy Putnam and H. this effect involves the regulation of the transmembrane movement and
Houston Merritt, when they discovered that it could be used to control intracellular distribution of Na+, K+, and Ca2+ (Keppel Hesselink,
seizures, while avoiding the sedation that was typical to phenobarbital 2017; Martin et al., 2014). Phenytoin has an especially significant
(Merritt and Putnam, 1984). Phenytoin has been examined and eval- blocking effect on the Na+ channels of neurons with high-frequency
uated in the context of many illnesses subsequent to its original epilepsy abnormal discharge, where it inhibits these repeated high-frequency
indication. For several disorders, phenytoin is still undergoing some discharges; in contrast, phenytoin has no significant effect on normal
form of innovation, including for bipolar disorder and wound healing, low-frequency discharges (Martin et al., 2014). In addition, phenytoin
amongst others (Keppel Hesselink, 2018). Using meta-analysis, it has inhibits the rapidly inactivating (T-type) Ca2+ channels of neurons and
also been revealed that phenytoin may be used to manage different inhibits Ca2+ influx (Keppel Hesselink and Kopsky, 2017a). Through
types of ulcers (Thangaraju et al., 2015). The use of phenytoin as a this mechanism, phenytoin affects the transmission of Ca2+-dependent
neuroprotector, in the context of multiple sclerosis, has also been ex- multiple neurotransmitters at the synapse, and the post-synaptic re-
plored in clinical trials (Raftopoulos et al., 2016). sponses.
Phenytoin also regulates hormone secretion of endocrine glands
cells and stimulates metabolism in the liver drug enzyme system.
3. Chemistry
Moreover, phenytoin increases high density lipoprotein concentration,
and stimulates granulation and capillary formation in healing tissues
Phenytoin (Fig. 1) is a hydantoin derivative (5,5-difenylhydantoine,
(Taing et al., 2017; Hesselink, 2017). The stability of phenytoin effects
5,5′-diphenylimidazolidine-2,4-dione; C15H12N2O2; M.W. 252.27; CAS
on cell membrane bioelectric activity is manifested in its ability to
Registry Number 57-41-0). It is a fine white or almost white crystalline
correct excessive cell excitement caused by various activities: phenytoin
powder with the following characteristics: m.p. 295–298 °C, (O'Neil,
can correct the excessive excitability of squid giant axonal cells caused
2001); pKa = 8.33, (Sangster, 1994); log P (octanol water) = 2.47,
by high-frequency electrical stimulation or low Ca2+ and Mg2+; phe-
(Hansch et al., 1995); slightly soluble in water (32 mg/L), (Yalkowsky
nytoin can increase resting potential in a high-potassium depolarisation
and He, 2003). Moreover, phenytoin is soluble in alkali hydroxides,
state, and reduce the frequency of endplate electrical sobbing, thereby
alcohol, acetone (O'Neil, 2001) and acetic acid (Lide and Milne, 1994).
affecting the motor nerve endplate potential (Suwalsky et al., 2006). In
Phenytoin is sensitive to light (Sunshine, 1969) and solutions are yel-
a recent study, phenytoin was shown to inhibit Ca2+ in CD38 pathways
lowed in light (Thompson and Micromedex, 2007). Finally, phenytoin
and cause oxidative stress, inflammation, and depression (Sadeghi
is odourless (or almost odourless) and tasteless (IARC, 1972).
et al., 2018).
There are two methods through which phenytoin can be synthe-
Phenytoin regulates a series of neurotransmitters including acet-
sised. Phenytoin synthesis can be achieved by adding base-catalysed
ylcholine, serotonin, norepinephrine, dopamine, GABA, and endorphins
urea to benzyl to produce benzilic acid, from which phenytoin is pro-
(Chou et al., 2014; Granger et al., 1995). At high concentrations, phe-
duced following a rearrangement. This process is called Biltz synthesis
nytoin can inhibit GABA uptake at nerve endings, indirectly enhancing
of phenytoin (Safari et al., 2009). In addition, phenytoin can be
the role of GABA by inducing GABA receptor proliferation; the end
result is an increase in Cl− influx and hyperpolarisation, and sub-
sequent inhibition of the incidence and spread of abnormal high-fre-
quency discharge (Chou et al., 2014; Zhang et al., 2019). While phe-
nytoin also reduces the concentration of acetylcholine in the brain,
small doses of phenytoin promote the release of acetylcholine from the
intestinal parasympathetic nerve terminals and ganglia, thereby sti-
mulating contraction of the Gastro-intestinal tract (Zhang et al., 2019).
In cardiac tissue, phenytoin shortens cardiac action potentials and
prolongs the refractory period (Razmaraii et al., 2016). In the central
nervous system, phenytoin targets neurons with high-frequency ac-
tivity, though most of its actions are exerted on the motor cortex (Chao
and Alzheimer, 1995). Thus, any spread from a seizure's focal point is
Fig. 1. Chemical structure of phenytoin. prevented and activity in the brain that is known to cause the tonic-

2
J. Patocka, et al. Food and Chemical Toxicology 142 (2020) 111393

clonic seizure's tonic phase is diminished (Osorio and Reed, 1989). 5.2. Side effects of phenytoin

Common side effects of phenytoin include a large number of non-


5. Pharmacokinetics specific symptoms (Hwang and Tsai, 2004). These include drowsiness
(Salinsky et al., 1996), fatigue (Siniscalchi et al., 2013), loss of control
5.1. Absorption, distribution, metabolism, and excretion (ADME) of bodily movements and loss of balance or coordination (Iivanainen
and Savolinen, 1983), irritability (Stephens and Shaffer, 1970), rest-
Phenytoin is well absorbed following oral administration (Urashima lessness (Drake, 1988), and several involuntary functions (Ahmad et al.,
et al., 2019). Its absorption takes place predominantly in the duo- 1975; Gunduz et al., 2013) including involuntary eye movements
denum. The rate of dissolution of phenytoin in the intestines informs its (Bittencourt et al., 1980; Matsue et al., 1981).
level of absorption. Peak plasma concentrations are typically reached Phenytoin can cause cutaneous adverse reactions, from maculo-
during a four to 8-h period. The distribution of the drug is widespread papular exanthema to SCAR (severe cutaneous adverse reaction), in-
throughout the body (distribution volume 0.8 L/kg). Phenytoin easily cluding drug reactions with eosinophilia, and systemic symptoms, in-
crosses the blood – brain barrier and it is extensively bound (~90%) to cluding Stevens-Johnson syndrome and toxic epidermal necrolysis. The
plasma albumin (Treiman and Woodbury, 1995). Phenytoin also easily pharmacogenomic basis of these severe cutaneous adverse side effects
crosses the placenta (Mirkin, 1971). Indeed, similar plasma con- can be linked to genetic factors (Chung et al., 2014).
centrations of phenytoin have been measured in maternal and umbilical Typically, the reactions include poor coordination, an increase in
cord (Nau et al., 1982). However, levels are typically low in breast milk the growth of hair, stomach aches, loss of appetite, nausea, and gingival
obtained from mothers with epilepsy (Mirkin, 1971). enlargement (Arya et al., 2012; Chacko and Abraham, 2014; Onaolapo
Phenytoin is metabolised by cytochrome P450 enzyme to 5-(p-hy- et al., 2018). Potentially more serious side effects include liver pro-
droxyphenyl)-5-phenylhydantoin (4′-HPPH). Further metabolisation to blems (Lee et al., 1976; Curry et al., 2018), bone marrow suppression
a catechol is possible, and this can extemporaneously oxidise to qui- (Sugimoto et al., 1982) and toxic epidermal necrolysis (Wu et al.,
none and semiquinone species (Cuttle et al., 2000; Ozkaynakci et al., 2018). Hepatotoxicity related to phenytoin is a serious idiosyncratic
2015). Phenytoin metabolism has been widely studied in vitro with side effect that is observed in a small percentage of those undergoing
respect to human subjects (Yasumori et al., 1999; Komatsu et al., 2000) treatment. It is accompanied by a host of symptoms ranging from a
and in vivo (Maguire, 1988; Szabo et al., 1990). Four oxidative meta- fever to arthralgias (Smythe and Umstead, 1989). In some patients,
bolites are reported, 4′-HPPH, 3′-HPPH, 3′,4′-diHPPH, and 3′,4′-dihy- phenytoin can cause liver necrosis (Lee et al., 1976).
drodiol (Maguire, 1988; Szabo et al., 1990). Research suggests that Many other syndromes have been attributed to phenytoin use. These
phenytoin is oxidised to 4′-HPPH by CYP2C9 in humans, and to a minor include: paradoxical seizure (Chua et al., 1999); drug withdrawal sei-
extent by CYP2C19 (Bajpai et al., 1996). However, the role played by zure (Nolan et al., 2013a,b); iplopia, blurred vision, and colour dis-
the P450s in the development of other metabolites requires further turbances (López et al., 1999); encephalopathy (Mehndiratta, 2016);
investigation. Recent reports suggest that the contribution of CYP3A4, ventricular fibrillation (Merlo González et al., 2002); atrioventricular
CYP2C9, and CYP2C19 to liver microsomal 3′,4′-diHPPH formation conduction disorder (Magadle et al., 1999); enlarged lymph nodes
from primary hydroxylated metabolites differs according to the in- (Schwinghammer and Howrie, 1983); purple spots on the skin (Oelze
dividual person (Komatsu et al., 2000; Lakehal et al., 2002). CYP2C9 and Pillow, 2013); and maculopapular exanthema leading to severe
variants are observed to play a role in the metabolism of phenytoin in cutaneous adverse side effects. These are supplemental to the common
the Chinese population (Chen et al., 2016). Furthermore, according to side effects described earlier, such as systematic and eosinophilia
recent studies, phenytoin has much higher CYP3A4-inducing potency. symptoms, toxic epidermal necrolysis, Stevens-Johnson syndrome
Therefore, CYP3A4-metabolised drugs may generally be required (Chung et al., 2014), coarsening of facial features (Sasaki et al., 1999),
during concurrent treatment with phenytoin (Hole et al., 2018). periarteritis nodosa (Haas, 1967), megaloblastic (folate-deficiency)
Research has also been conducted to study phenytoin metabolism in anaemia (Scott and Weir, 1980), low blood calcium levels (hypo-
animal subjects (Chow et al., 1980; Billings, 1983; Doecke et al., 1990). calcaemia) (Nseir et al., 2013), and immunoglobulin abnormalities
It should be noted that rates of 4′-HPPH formation in rat liver micro- (Yabuki and Nakaya, 1976).
somes were 10 times higher than in humans (Munns et al., 1997). The In addition, adverse effects related with phenytoin use can include
reasons for this difference in P450 metabolism are still unclear. Phe- various psychiatric disorders or behavioural symptoms, specifically
nytoin has been shown to induce several forms of P450 (Fleishaker those associated with vitamin B12 or a folic acid deficiency (Reynolds,
et al., 1995; Ghosal et al., 1996; Yamazaki et al., 1996). However, it is 1967; Matsuura, 1999; Gatzonis et al., 2003). A rare case of toxicity was
unknown whether administrating phenytoin can lead to auto-induced recently reported that presented itself as psychosis resulting from vi-
metabolism in humans and rats (Edeki and Brase, 1995; Chetty et al., tamin deficiency (Borasi et al., 2015). However, it is necessary to re-
1998). cognise that psychological and behavioural side effects occur more
Phenytoin displays non-linear elimination pharmacokinetics frequently in patients taking antiepileptics other than phenytoin (Chen
(Browne and LeDuc, 1995). A result of this non-linearity is that the et al., 2017).
elimination half-life differs with plasma concentration. A saturation of
metabolite-inducing enzymes is the reason for the non-linearity in the 5.3. DRESS syndrome
elimination of phenytoin (Browne and LeDuc, 1995). As shown in
Fig. 2, the main primary metabolites are 5(3′-hydroxyphenyl)-5-phe- Allergic reactions to phenytoin present themselves as a rash. They
nylhydantoin (3′-HPPH), 5(4′-hydroxyphenyl)-5-phenylhydantoin (4′- can sometimes, if rarely, take up more severe forms, e.g. anaphylaxis or
HPPH), 5(3′,4′-dihydroxyphenyl)-5-phenylhydantoin (3′,4′-diHPPH), DRESS (drug reaction with eosinophilia and systemic symptoms)
and 5(3′,4′-dihydroxy-1′,5′-cyclohexydiene-1-yl)-5-phenylhydantoin (Kumkamthornkul et al., 2018). A side effect linked to DRESS syndrome
(3′,4′-dihydrodiol). In addition, arene-oxide is a highly reactive and is the drug-induced hypersensitivity syndrome (amongst other names).
extremely unstable hypothetical metabolite which has never been iso- This side effect is a known SCAR (severe cutaneous adverse reaction)
lated. Arene oxide may be responsible for the carcinogenic effects of and can prove to be life-threatening (Sullivan and Shear, 2001). Drug-
phenytoin. The main secondary metabolites are glucuronide con- induced hypersensitivity syndrome is associated with phenytoin in most
jugates of phenytoin and its hydroxylated derivatives (Scriba et al., cases and was originally referred to as phenytoin hypersensitivity
1995). syndrome (De et al., 2018). Although this syndrome was subsequently
discovered to occur with several different drugs, phenytoin remains a

3
J. Patocka, et al. Food and Chemical Toxicology 142 (2020) 111393

Fig. 2. Proposed pathways of phenytoin metabolism.

frequent cause of this allergic reaction (Brizendine and Naik, 2013; CNS dysfunction, viz. ataxia, drowsiness, and nystagmus (Browne,
Deka et al., 2013; Hall and Fromm, 2013; Velasco and McDermott, 1997). However, these reactions can be reversed when the dosage is
2014; Yampayon et al., 2017). DRESS typically has a two to six-week reduced. A chronic overdose has been found to be responsible for cer-
latency period. While it is known that the pathophysiology of DRESS ebellar atrophy in adult patients being administered phenytoin
involves the activation of lymphocytes and reactivation of viruses, its (Kokenge et al., 1965; Selhorst et al., 1972; Ghatak et al., 1976; Mc Lain
pathophysiology is not completely understood (Yampayon et al., 2017). et al., 1980; Baier et al., 1984; Ney et al., 1994; Ahuja et al., 2000; Tan
DRESS is normally associated with a morbilliform cutaneous eruption, et al., 2001; De Marcos et al., 2003; Lee et al., 2003; Chow and Szeto,
and this is accompanied by lymphadenopathy, and a rise in tempera- 2007; Kumar et al., 2013). Evidence has been reported that CYP2C9
ture. The severity of DRESS is linked to systemic involvement, and it polymorphisms are associated with an increase in the frequency of
can result in multiple organ-failure (Ghannam et al., 2017). cerebellar atrophy following the use of phenytoin (Silvado et al., 2018).

5.4. Cerebellar atrophy 6. Toxicology

The adverse side effects of phenytoin can be chronic and/or acute. The therapeutic range for phenytoin is narrow and a total serum
As the amount of phenytoin rises, there is a disproportionate rise in the concentration greater than 80 μM is, for many patients, linked to
plasma drug concentration, despite the increments in dosage itself clinically relevant toxicity (Praveen-kumar and Desai, 2014). Phenytoin
being small, and this increase in plasma drug concentration is accom- is typically excreted by the kidneys after it has been metabolised by
panied by a change from subtherapeutic to toxic levels (McNamara, hepatic enzymes. However, a toxic accumulation of phenytoin can
1996). These plasma drug concentration changes are highly linked to present itself in the context of people with renal failure (Chua et al.,

4
J. Patocka, et al. Food and Chemical Toxicology 142 (2020) 111393

Table 1
Published toxicity data for phenytoin.
Species Test Typea Route Reported Dose Source

mouse LD50 intravenous 92 mg/kg Stille and Brunckow (1954)


mouse LD50 intraperitoneal 100 mg/kg TOXNET
mouse LD50 subcutaneous 110 mg/kg Stille and Brunckow (1954)
mouse LD50 Oral 150 mg/kg TOXNET
mouse LD50 unreported 800 mg/kg (800 mg/kg) TOXNET
rat LD50 intravenous 101 mg/kg TOXNET
rat LD50 intraperitoneal 352 mg/kg Kemp et al. (1971)
rat LD50 oral 1635 mg/kg Graziani et al. (1983)
rat LD50 subcutaneous > 1500 mg/kg Masuda et al. (1980)
rabbit LD50 intravenous 56.4 mg/kg Graziani et al. (1983)
rabbit LD50 oral > 3000 mg/kg TOXNET
dog LD50 intravenous 90 mg/kg Usdin and Efron (1972)
child LDLo oral 100 mg/kg Theil et al. (1961)
child TDLo intravenous 15 mg/kg Howrie and Crumrine (1985)
child TDLo intravenous 15 mg/kg Krishnamoorthy et al. (1983)
child TDLo oral 3 mg/kg Wilson et al. (1976)
child TDLo oral 11 mg/kg Zinsmeister and Marks. (1976)
child LDLo oral 140 mg/kg Bajoghli (1961)
child TDLo unreported 18 mg/kg Zinsmeister and Marks (1976)
man TDLo oral 31 mg/kg TOXNET
man TDLo oral 1300 mg/kg Gams et al. (1968)
women TDLo oral 106 mg/kg TOXNET
women TDLo oral 200 mg/kg Weichbrodt and Elliott, 1987.
women TDLo oral 540 mg/kg Mahatma et al. (1989)

a
LD50 = Median Lethal Dose, LDLo = Lethal Dose Low, TDLo = Toxic Dose Low, NOAEL = No-Observed-Adverse-Effect Level.

2000). Haemodialysis does not remove phenytoin because it is 90% (NTP, 1993). Hepatocellular neoplasia in phenytoin-treated rodents
albumin bound (Martin et al., 1977). While this form of toxicity is not have little to no relevance for humans. At present, in the context of
typically fatal, it can lead to neurological symptoms ranging from a epilepsy patients, no link has been found between cancer of the liver
coma (Craig, 2005), to nystagmus (Praveen-kumar and Desai, 2014), to and phenytoin (Dethloff et al., 1996; Singh et al., 2005; Friedman et al.,
ataxia (Shanmugarajah et al., 2018). On rare occasions, intravenous 2009).
applications may face complications stemming from Purple Glove There are some indications that phenytoin is linked to a larger risk
Syndrome (PGS) (Garbovsky et al., 2015). PGS is a serious side effect of congenital malformations (Danielsson et al., 2005; Eroğlu et al.,
that can lead to amputation (Jaain et al., 2015; Okogbaa et al., 2015). 2008). The risk of congenital malformations is linked to phenytoin's
Toxicity relating to phenytoin can be a product of dose misuse, dose antiepileptic activity. A good number of studies have revealed that this
adjustment, an interaction with other drugs, or a patient's changing is the main risk factor for a rise in the incidence of congenital mal-
physiology (Kang et al., 2013; Srinivasan et al., 2015). The first formations. The risks associated with teratogenicity are multifactorial
symptoms of phenytoin intoxication are nausea, dysfunctions related to and include factors such as a patient's genetic predisposition (Canger
the central nervous system (such as ataxia or nystagmus), a depressed et al., 1999).
state, and coma (Craig, 2005). Problems such as hypotension or ar- Typically, therapy against phenytoin toxicity would involve sup-
rhythmia are not typically linked to the use of this drug. However, in portive care. Therapy is focussed on the vital functions of the patient's
cases of parenteral administration, these problems may present them- body, alongside managing vomit and nausea. In addition, supportive
selves. Phenytoin is considered a moderately toxic substance (Imam care is essential to prevent injuries that can otherwise result from ataxia
et al., 2014). On its own, the likelihood that phenytoin intoxication will or confusion. There is no antidote to phenytoin toxicity, and little
result in patient death is considered to be very low. The published acute evidence to suggest that enhanced elimination or gastrointestinal de-
toxicity data for phenytoin in different organisms are summarised in contamination can speed up recovery (Zhang et al., 2019).
Table 1. Data on mutagenicity, carcinogenicity, and teratogenicity are
provided in separate subchapters.
7. Conclusions
The mutagenicity of phenytoin and the mutagenicity of its major
metabolite, 5-(4-hydroxyphenyl)-5-phenylhydantoin (HPPH), have
Phenytoin is a long-standing antiepileptic drug which exerts its ef-
been tested in vitro on different Salmonella typhimurium strains (TA1535,
fects by reducing the influx of Na+ and Ca2+ into nerve cells and
TA100, TA1537, TA1538, TA98) using an Ames test (Sezzano et al.,
promoting the release of GABA; thus phenytoin effectively inhibits the
1982; Léonard et al., 1984; Riedel and Obe, 1984). From the results of
abnormal discharge of neurons, prevents the transmission of abnormal
these studies, it was concluded that phenytoin does not have mutagenic
impulses, and reduces the symptoms of withdrawal. In the brain, opioid
properties.
μ and δ receptors mediate epilepsy-like reactions, and endogenous
The experimental approach used in earlier mutagenicity tests
opioid peptides and opioid receptors are involved in the pathophy-
(Montes de Oca-Luna et al., 1984; Barcellona et al., 1987) is likely to be
siology of primary epilepsy. A correlation may exist between the pa-
responsible for generating reactive arene-oxide (Daly et al., 1972), an
thogenesis of addictive diseases and epilepsy. Therefore, antiepileptic
extremely unstable metabolite that has never been isolated (Brown and
drugs may be used to treat addictive diseases. Phenytoin treatment can
LeDuc, 1995). Therefore, phenytoin was found not to be directly gen-
control withdrawal symptoms in heroin addicts and can effectively
otoxic in the context of a set of shorter-term assays using cytogenetics
reduce the use of alternatives, such as methadone, in the treatment of
(Kindig et al., 1992).
opioid addicts without affecting their efficacy. Moreover, phenytoin
Experiments in laboratory animals have not provided any evidence
effectively improves symptoms of anxiety, craving, and protracted
of phenytoin carcinogenicity (Jang et al., 1987; Maeda et al., 1988;
withdrawal.
Chhabra et al., 1993); although in one case the results are inconsistent
Phenytoin-induced toxicity can occur following an increase in the

5
J. Patocka, et al. Food and Chemical Toxicology 142 (2020) 111393

daily dose, a change in formulation or brand, or a change in the fre- Press, New York, pp. 283–300.
quency of administration. In recent years, there have been many reports Canger, R., Battino, D., Canevini, M.P., Fumarola, C., Guidolin, L., Vignoli, A., Mamoli, D.,
Palmieri, C., Molteni, F., Granata, T., Hassibi, P., Zamperini, P., Pardi, G., Avanzini,
of adverse reactions caused by therapeutic doses of phenytoin. G., 1999. Malformations in offspring of women with epilepsy: a prospective study.
However, there is no specific antidote to phenytoin toxicity. Therefore, Epilepsia 40 (9), 1231–1236.
individualised administration is of great significance for the safe and Chacko, L.N., Abraham, S., 2014. Phenytoin-induced gingival enlargement. BMJ Case
Rep. 2014 pii: bcr2014204670.
effective use of phenytoin. With the widespread application of new Chao, T.I., Alzheimer, C., 1995. Effects of phenytoin on the persistent Na+ current of
molecular biology techniques, current research on individualised phe- mammalian CNS neurones. Neuroreport 6 (13), 1778–1780.
nytoin medication has gradually evolved from traditional drug therapy Chen, L.G., Wang, Z., Zhu, Y., Xiong, J.H., Sun, L.R., Dai, D.P., Cai, J.P., Hu, G.X., 2016. In
vitro metabolism of phenytoin in 36 CYP2C9 variants found in the Chinese popula-
monitoring to pharmacogenomics. From population pharmacokinetics, tion. Chem. Biol. Interact. 253, 93–99.
nonlinear mixed effect modelling (NONMEM) has led to the develop- Chen, B., Choi, H., Hirsch, L.J., Katz, A., Legge, A., Buchsbaum, R., Detyniecki, K., 2017.
ment of a useful tool for designing gene-oriented and individualised Psychiatric and behavioral side effects of antiepileptic drugs in adults with epilepsy.
Epilepsy Behav. 76, 24–31.
drug regimens.
Chetty, M., Miller, R., Seymour, M.A., 1998. Phenytoin auto-induction. Ther. Drug Monit.
In summary, phenytoin has a wide range of biological activities and 20, 60–62.
pharmacological indications. In addition to being used clinically to Chhabra, R.S., Bucher, J.R., Haseman, J.K., Elwell, M.R., Kurtz, P.J., Carlton, B.D., 1993.
treat epilepsy, phenytoin is also used to treat a variety of diseases, in- Comparative carcinogenicity of 5,5-diphenylhydantoin with or without perinatal
exposure in rats and mice. Fund. Appl. Toxicol. 21 (2), 174–186 Erratum in: Fundam
cluding migraine, dizziness, hiccups, myocardial infarction, burns, and Appl Toxicol 1994; 22(1): 159.
in promoting wound healing. Although phenytoin has been used in Chou, M.Y., Lee, C.Y., Liou, H.H., Pan, C.Y., 2014. Phenytoin attenuates the hyper-ex-
epilepsy treatment for many years and has been somewhat superseded citing neurotransmission in cultured embryonic cortical neurons.
Neuropharmacology 83, 54–61.
by newer anti-epileptic medicines, there is good reason to believe that Chow, K.M., Szeto, C.C., 2007. Cerebral atrophy and skull thickening due to chronic
phenytoin will find new life as a drug for the individualised treatment phenytoin therapy. CMAJ (Can. Med. Assoc. J.) 176 (3), 321–323.
of epilepsy, addiction, migraine, and other diseases. Chow, S.A., Charkowski, D.M., Fischer, L.J., 1980. Separation of phenytoin and its me-
tabolites by high-performance liquid chromatography. Life Sci. 27 (25–26),
2477–2482.
Declaration of competing interest Chua, H.C., Venketasubramanian, N., Tan, C.B., Tjia, H., 1999. Paradoxical seizures in
phenytoin toxicity. Singap. Med. J. 40 (4), 276–277.
Chua, H.C., Venketasubramanian, N., Tjia, H., Chan, S.P., 2000. Elimination of phenytoin
The authors declare that they have no known competing financial in toxic overdose. Clin. Neurol. Neurosurg. 102 (1), 6–8.
interests or personal relationships that could have appeared to influ- Chung, W.H., Chang, W.C., Lee, Y.S., Wu, Y.Y., Yang, C.H., Ho, H.C., Chen, M.J., Lin, J.Y.,
Hui, R.C., Ho, J.C., Wu, W.M., Chen, T.J., Wu, T., Wu, Y.R., Hsih, M.S., Tu, P.H.,
ence the work reported in this paper.
Chang, C.N., Hsu, C.N., Wu, T.L., Choon, S.E., Hsu, C.K., Chen, D.Y., Liu, C.S., Lin,
C.Y., Kaniwa, N., Saito, Y., Takahashi, Y., Nakamura, R., Azukizawa, H., Shi, Y.,
Acknowledgement Wang, T.H., Chuang, S.S., Tsai, S.F., Chang, C.J., Chang, Y.S., Hung, S.I., 2014.
Taiwan severe cutaneous adverse reaction consortium; Japan pharmacogenomics
data science consortium. Genetic variants associated with phenytoin-related severe
This work was supported by the Excelence project UHK. cutaneous adverse reactions. J. Am. Med. Assoc. 312 (5), 525–534.
Craig, S., 2005. Phenytoin poisoning. Neurocritical Care 3 (2), 161–170.
References Curry, B., Mican, L., Smith, T.L., 2018. Phenytoin-induced chronic liver enzyme elevation
and hepatic fibrosis: a case report. Ment Health Clin 8 (4), 184–187.
Cuttle, L., Munns, A.J., Hogg, N.A., Scott, J.R., Hooper, W.D., Dickinson, R.G., Gillam,
Abou-Khalil, B.W., 2016. Antiepileptic Drugs. Continuum (Minneap Minn). 22. pp. E.M., 2000. Phenytoin metabolism by human cytochrome P450: involvement of P450
132–156. 3A and 2C forms in secondary metabolism and drug-protein adduct formation. Drug
Ahmad, S., Laidlaw, J., Houghton, G.W., Richens, A., 1975. Involuntary movements Metab. Dispos. 28 (8), 945–950.
caused by phenytoin intoxication in epileptic patients. J. Neurol. Neurosurg. Daly, J.W., Jerina, D.M., Witkop, B., 1972. Arene oxides and the NIH shift: the metabo-
Psychiatry 38 (3), 225–231. lism, toxicity and carcinogenicity of aromatic compounds. Experientia 28 (10),
Ahuja, S.R., Karande, S., Kulkarni, M.V., 2000. Cerebellar atrophy in an epileptic child: is 1129–1149.
it due to phenytoin? J. Postgrad. Med. 46 (4), 278–279. Danielsson, B.R., Johansson, A., Danielsson, C., Azarbayjani, F., Blomgren, B., Sköld, A.C.,
Arya, R., Gulati, S., 2012. Phenytoin-induced gingival overgrowth. Acta Neurol. Scand. 2005. Phenytoin teratogenicity: hypoxia marker and effects on embryonic heart
125 (3), 149–155. rhythm suggest an hERG-related mechanism. Birth Defects Res Clin Mol Teratol 73
Ashnagar, A., Gharib Naseri, N., Amini, M., 2009. Synthesis of 5,5-diphenyl-2,4-imisda- (3), 146–153.
zolidinedione (phenytoin) from almond. Asian J. Chem. 21 (7), 4976–4980. De, A., Rajagopalan, M., Sarda, A., Das, S., Biswas, P., 2018. Drug reaction with eosi-
Baier, W.K., Beck, U., Doose, H., Klinge, H., Hirsch, W., 1984. Cerebellar atrophy fol- nophilia and systemic symptoms: an update and review of recent literature. Indian J.
lowing diphenylhydantoin intoxication. Neuropediatrics 15 (2), 76–81. Dermatol. 63 (1), 30–40.
Bajoghli, M., 1961. Generalized lymphadenopathy and hepatosplenomegaly induced by De Marcos, F.A., Ghizoni, E., Kobayashi, E., Li, L.M., Cendes, F., 2003. Cerebellar volume
diphenylhydantoin. Pediatrics 28, 943–945. and long-term use of phenytoin. Seizure 12 (5), 312–315.
Bajpai, M., Roskos, L.K., Shen, D.D., Levy, R.H., 1996. Roles of cytochrome P4502C9 and Deka, N.M., Dass, R., Das, B.K., Hoque, R., 2013. Phenytoin induced DRESS syndrome.
cytochrome P4502C19 in the stereoselective metabolism of phenytoin to its major Indian J. Pediatr. 80 (3), 266.
metabolite. Drug Metab. Dispos. 24 (12), 1401–1403. Dethloff, L.A., Graziano, M.J., Goldenthal, E., Gough, A., de la Iglesia, F.A., 1996.
Barcellona, P.S., Barale, R., Campana, A., Zucconi, D., Rossi, V., Caranti, S., 1987. Perspective on the carcinogenic potential of phenytoin based on rodent tumor
Correlations between embryotoxic and genotoxic effects of phenytoin in mice. bioassays and human epidemiological data. Hum. Exp. Toxicol. 15 (4), 335–348.
Teratog. Carcinog. Mutagen. 7 (2), 159–168. Doecke, C.J., Sansom, L.N., McManus, M.E., 1990. Phenytoin 4-hydroxylation by rabbit
Barratt, E.S., Stanford, M.S., Felthous, A.R., Kent, T.A., 1997. The effects of phenytoin on liver P450IIC3 and identification of orthologs in human liver microsomes. Biochem.
impulsive and premeditated aggression: a controlled study. J. Clin. Psychopharmacol. Biophys. Res. Commun. 166, 860–866.
17 (5), 341–349. Drake, M.E., 1988. Restless legs with antiepileptic drug therapy. Clin. Neurol. Neurosurg.
Billings, R.E., 1983. Sex differences in rats in the metabolism of phenytoin to 5-(3,4- 90 (2), 151–154.
dihydroxyphenyl)-5-phenylhydantoin. J. Pharmacol. Exp. Therapeut. 225, 630–636. Edeki, T.I., Brase, D.A., 1995. Phenytoin disposition and toxicity: role of pharmacogenetic
Bittencourt, P.R., Gresty, M.A., Richens, A., 1980. Quantitative assessment of smooth- and interethnic factors. Drug Metab. Rev. 27, 449–469.
pursuit eye movements in healthy and epileptic subjects. J. Neurol. Neurosurg. Eroğlu, E., Gökçil, Z., Bek, S., Ulaş, U.H., Odabaşi, Z., 2008. Pregnancy and teratogenicity
Psychiatry 43 (12), 1119–1124. of antiepileptic drugs. Acta Neurol. Belg. 108 (2), 53–57.
Borasi, M., Verma, R.P., Gupta, S.K., 2015. Psychosis as harbinger of phenytoin toxicity. Farook, F.F., Nizam, M.N.M., Alshammari, A., 2019. An update on the mechanisms of
Toxicol. Int. 22 (1), 160–161. phenytoin induced gingival overgrowth. Open Dent. J. 13, 430–435.
Bosch, J., Roca, T., Domènech, J., Suriol, M., 1999. Synthesis of water-soluble phenytoin Fleishaker, J.C., Pearson, L.K., Peters, G.R., 1995. Phenytoin causes a rapid increase in 6β-
prodrugs. Bioorg. Med. Chem. Lett 9 (13), 1859–1862. hydroxycortisol urinary excretion in humans—a putative measure of CYP3A induc-
Brizendine, C.E., Naik, P.J., 2013. Drug rash with eosinophilia and systemic symptoms tion. J. Pharmaceut. Sci. 84, 292–294.
syndrome associated with use of phenytoin, divalproex sodium, and phenobarbital. Friedman, G.D., Jiang, S.F., Udaltsova, N., Quesenberry Jr., C.P., Chan, J., Habel, L.A.,
Am. J. Health Syst. Pharm. 70 (6), 507–510. 2009. Epidemiologic evaluation of pharmaceuticals with limited evidence of carci-
Browne, T.R., 1997. Phenytoin and other hyadantoins. In: Engel Jr.J., Pidley, T. (Eds.), nogenicity. Int. J. Canc. 125 (9), 2173–2178.
Epilepsy. A Comprehensive Textbook, first ed. Lippincott-Raven, Philadelphia, pp. Gams, R.A., Neal, J.A., Conrad, F.G., 1968. Hydantoin-induced pseudo-pseudolymphoma.
1557–1579. Ann. Intern. Med. 69 (3), 557–568.
Browne, T.R., LeDuc, B., 1995. Phenytoin: chemistry and biotransformation. In: Levy, Garbovsky, L.A., Drumheller, B.C., Perrone, J., 2015. Purple glove syndrome after phe-
R.H., Mattson, R.H., Meldrum, B.S. (Eds.), Antiepileptic Drugs, fourth ed. Raven nytoin or fosphenytoin administration: review of reported cases and

6
J. Patocka, et al. Food and Chemical Toxicology 142 (2020) 111393

recommendations for prevention. J. Med. Toxicol. 11 (4), 445–459. identify phenytoin and carbamazepine provoked DRESS or SJS/TEN in epilepsy pa-
Gatzonis, S.D., Angelopoulos, E., Sarigiannis, P., Mantouvalos, V., Ploumbidis, D., tients. Int. Immunopharm. 63, 204–210.
Siafakas, A., 2003. Acute psychosis due to treatment with phenytoin in a nonepileptic Lakehal, F., Wurden, C.J., Kalhorn, T.F., Levy, R.H., 2002. Carbamazepine and oxcar-
patient. Epilepsy Behav. 4 (6), 771–772. bazepine decrease phenytoin metabolism through inhibition of CYP2C19. Epilepsy
Ghannam, M., Mansour, S., Nabulsi, A., Abdoh, Q., 2017. Anticonvulsant hypersensitivity Res. 52 (2), 79–83.
syndrome after phenytoin administration in an adolescent patient: a case report and Lee, T.J., Carney, C.N., Lapis, J.L., Higgins, T., Fallon, H.J., 1976. Diphenylhydantoin-
review of literature. Clin. Mol. Allergy 15, 14. induced hepatic necrosis. A case study. Gastroenterology 70 (3), 422–424.
Ghatak, N.R., Santoso, R.A., McKinney, W.M., 1976. Cerebellar degeneration following Lee, S.K., Mori, S., Kim, D.J., Kim, S.Y., Kim, S.Y., Chu, M., Heo, K., Lee, B.I., Kim, D.I.,
long term phenytoin therapy. Neurology 26 (9), 818–820. 2003. Diffusion tensor MRI and fiber tractography of cerebellar atrophy in phenytoin
Ghosal, A., Sadrieh, N., Reik, L., Levin, W., Thomas, P.E., 1996. Induction of the male- users. Epilepsia 44 (12), 1536–1540.
specific cytochrome P450 3A2 in female rats by phenytoin. Arch. Biochem. Biophys. Léonard, A., de Meester, C., Fabry, L., de Saint-Georges, L., Dumont, P., 1984. Lack of
332, 153–162 1996. mutagenicity of diphenylhydantoin in in vitro short-term tests. Mutat. Res. 137 (2–3),
Granger, P., Biton, B., Faure, C., 1995. Modulation of the gammaaminobutyric acid type A 79–88.
receptor by the antiepileptic drugs carbamazepine and phenytoin. Mol. Pharmacol. Lewis, R.J., 2004. Sr. Sax's Dangerous Properties of Industrial Materials, eleventh ed.
47 (6), 1189–1196. Wiley-Interscience, Wiley & Sons, Inc., Hoboken, NJ, pp. 1302.
Graziani, G., Barbadoro, E., Testa, R., 1983. Denzimol, a new anticonvulsant drug. III. Handbook of Data on Organic Compounds, third ed. Lide, D.R., Milne, G.W.A. (Eds.),
Toxicological evaluation. Arzneimittelforschung 33 (8), 1168–1173. 1994. vol. 4. CRC Press, Inc., Boca Raton, FL, pp. 3216 vol. I.
Gunduz, T., Kocasoy-Orhan, E., Hanagasi, H.A., 2013. Orolingual dyskinesia and in- López, L., Thomson, A., Rabinowicz, A.L., 1999. Assessment of colour vision in epileptic
voluntary neck movements caused by phenytoin intoxication. J. Neuropsychiatry patients exposed to single-drug therapy. Eur. Neurol. 41 (4), 201–205.
Clin. Neurosci. 25 (4), E51. Maeda, T., Sano, N., Togei, K., Shibata, M., Izumi, K., Otsuka, H., 1988. Lack of carci-
Haas, P., 1967. [On a very acutely developing case of periarteritis nodosa following di- nogenicity of phenytoin in (C57BL/6 x C3H)F1 mice. J. Toxicol. Environ. Health 24
phenylhydantoin therapy]. Wien Klin. Wochenschr. 79 (3), 56–58 German. (1) 111-1119.
Hains, B.C., Saab, C.Y., Lo, A.C., Waxman, S.G., 2004. Sodium channel blockade with Magadle, R., Weiner, P., Waizman, J., 1999. [Atrioventricular junctional arrhythmia due
phenytoin protects spinal cord axons, enhances axonal conduction, and improves to oral phenytoin intoxication]. Harefuah 136 (6), 446–448 514. Hebrew.
functional motor recovery after contusion SCI. Exp. Neurol. 188 (2), 365–377. Maguire, J.H., 1988. Quantitative estimation of catechol/methylcatechol pathways in
Hall, D.J., Fromm, J.S., 2013. Drug reaction with eosinophilia and systemic symptoms human phenytoin metabolism. Epilepsia 29 (6), 753–759.
syndrome in a patient taking phenytoin and levetiracetam: a case report. J. Med. Case Mahatma, M., Haponik, E.F., Nelson, S., Lopez, A., Summer, W.R., 1989. Phenytoin-in-
Rep. 7, 2. duced acute respiratory failure with pulmonary eosinophilia. Am. J. Med. 87 (1),
Hall, E.A., Wheless, J.W., Phelps, S.J., 2020. Status epilepticus: the slow and agonizing 93–94.
death of phenytoin. J. Pediatr. Pharmacol. Therapeut. 25 (1), 4–6. Martin, E., Gambertoglio, J.G., Adler, D.S., Tozer, T.N., Roman, L.A., Grausz, H., 1977.
Hansch, C., Leo, A., Hoekman, D., 1995. Exploring QSAR - Hydrophobic, Electronic, and Removal of phenytoin by hemodialysis in uremic patients. J. Am. Med. Assoc. 238
Steric Constants. American Chemical Society., Washington, DC, pp. 129. (16), 1750–1753.
Hesselink, J.M.K., 2017. Phenytoin: a step by step insight into its multiple mechanisms of Martin, L.J., Chao, R., Corry, B., 2014. Molecular dynamics simulation of the partitioning
action—80 years of mechanistic studies in neuropharmacology. J. Neurol. 264 (9), of benzocaine and phenytoin into a lipid bilayer. Biophys. Chem. 185, 98–107.
2043–2047. Masuda, Y., Karasawa, T., Shiraishi, Y., Hori, M., Yoshida, K., Shimizu, M., 1980. 3-
Hole, K., Wollmann, B.M., Nguyen, C., Haslemo, T., Molden, E., 2018. Comparison of Sulfamoylmethyl-1,2-benzisoxazole, a new type of anticonvulsant drug.
CYP3A4-inducing capacity of enzyme-inducing antiepileptic drugs using 4β-hydro- Pharmacological profile. Arzneimittelforschung 30 (3) 477-4783.
xycholesterol as biomarker. Ther. Drug Monit. 40 (4), 463–468. Mathews, S.R., Badyal, D.K., Mathew, R., 2019. Phenytoin-induced bradycardia and hy-
Howrie, D.L., Crumrine, P.K., 1985. Phenytoin-induced movement disorder associated potension. Indian J. Pharmacol. 51 (2), 120–122.
with intravenous administration for status epilepticus. Clin. Pediatr. 24 (8), 467–469. Matsue, Y., Wagatsuma, S., Ito, K., Okuma, T., 1981. Smooth pursuit eye movements in
Hwang, W.J., Tsai, J.J., 2004. Acute phenytoin intoxication: causes, symptoms, mis- epileptics under antiepileptic medication. Tohoku J. Exp. Med. 134 (4), 341–349.
diagnoses, and outcomes. Kaohsiung J. Med. Sci. 20 (12), 580–585. Matsuura, M., 1999. Epileptic psychoses and anticonvulsant drug treatment. J. Neurol.
IARC, 1972. Monographs on the Evaluation of the Carcinogenic Risk of Chemicals to Neurosurg. Psychiatry 67 (2), 231–233.
Humans, vol. 66. World Health Organization, International Agency for Research on McLain, L.W., Martain, J.T., Allen, J.H., 1980. Cerebellar degeneration due to chronic
Cancer, Geneva, pp. 175. Available at: http://monographs.iarc.fr/ENG/ phenytoin therapy. Ann. Neurol. 7 (1), 18–23.
Classification/index.php. McNamara, J.O., 1996. Drugs effective in the therapy of the epilepsies. In: Hardman, J.G.,
Iivanainen, M., Savolainen, H., 1983. Side effects of phenobarbital and phenytoin during Limbird, L.E., Molinoff, P.B., Ruddon, R.W. (Eds.), Goodman and Gilman's the
long-term treatment of epilepsy. Acta Neurol. Scand. Suppl. 97, 49–67. Pharmacological Basis of Therapeutics, ninth ed. McGrawHill, New York, pp.
Imam, S.H., Landry, K., Kaul, V., Gambhir, H., John, D., Kloss, B., 2014. Free phenytoin 461–486.
toxicity. Am. J. Emerg. Med. 32 (10) 1301.e3-4. Mehndiratta, S., 2016. Phenytoin-induced encephalopathy in a child. Indian J.
Jain, R.S., Nagpal, K., Kumar, S., Prakash, S., Handa, R., 2015. Purple glove syndrome Pharmacol. 48 (4), 460–461.
occurring after oral administration of phenytoin in therapeutic doses: mechanism still Merlo González, V.E., Arbol Linde, F., Rodríguez, J., López Onega, P., 2002. [Ventricular
a dilemma. Am. J. Emerg. Med. 33 (1) 123.e5-6. fibrillation during intravenous administration of phenytoin]. Med. Int. 19 (3),
Jang, J.J., Takahashi, M., Furukawa, F., Toyoda, K., Hasegawa, R., Sato, H., Hayashi, Y., 158–159 Spanish.
1987. Long-term in vivo carcinogenicity study of phenytoin (5,5-diphenylhydantoin) Merritt, H.H., Putnam, T.J., 1984. Landmark article Sept 17, 1938: sodium diphenyl
in F344 rats. Food Chem. Toxicol. 25 (9), 697–702. hydantoinate in the treatment of convulsive disorders. By H. Houston Merritt and
Kang, C.K., Kim, M.K., Kim, M.J., Jun, K.I., Kim, K.W., Park, S.H., Song, K.H., 2013. Acute Tracy J. Putnam. J. Am. Med. Assoc. 251 (8), 1062–1067.
respiratory failure caused by phenytoin overdose. Korean J. Intern. Med. (Engl. Ed.) Mirkin, M.L., 1971. Diphenylhydantoin: placental transport, fetal localisation, neonatal
28 (6), 736–738. metabolism, and possible teratogenic effects. J. Pediatr. 78 (2), 329–337.
Kemp, J.W., Tannhauser, M., Swinyard, E.A., 1971. Effect of diphenylhydantoin onhex- Montes de Oca-Luna, R., Leal-Garza, C.H., Baca-Sevilla, S., Garza-Chapa, R., 1984. The
obarbital sleep time in mice and rats. Arch. Int. Pharmacodyn. Ther. 193 (1), 37–47. effect of diphenylhydantoin on the frequency of micronuclei in bone-marrow poly-
Keppel Hesselink, J.M., 2018. Phenytoin repositioned in wound healing: clinical experi- chromatic erythrocytes of mice. Mutat. Res. 141 (3–4), 183–187.
ence spanning 60 years. Drug Discov. Today 23 (2), 402–408. Munns, A.J., De Voss, J.J., Hooper, W.D., Dickinson, R.G., Gillam, E.M.J., 1997.
Keppel Hesselink, J.M., Kopsky, D.J., 2017a. Phenytoin: 80 years young, from epilepsy to Bioactivation of phenytoin by human cytochrome P450: characterization of the
breast cancer, a remarkable molecule with multiple modes of action. J. Neurol. 264, mechanism and targets of covalent adduct formation. Chem. Res. Toxicol. 10,
1617–1621. 1049–1058.
Keppel Hesselink, J.M., Kopsky, D.J., 2017b. Phenytoin: neuroprotection or neurotoxi- Nau, H., Kuhnz, W., Egger, H.J., Rating, D., Helge, H., 1982. Anticonvulsants during
city? Neurol. Sci. 38, 1137–1141. pregnancy and lactation. Transplacental, maternal and neonatal pharmacokinetics.
Kindig, D., Garriott, M.L., Parton, J.W., Brunny, J.D., Beyers, J.E., 1992. Clin. Pharmacokinet. 7 (6), 508–543.
Diphenylhydantoin is not genotoxic in a battery of short-term cytogenetic assays. Ney, G.C., Lantos, G., Barr, W.B., Schaul, N., 1994. Cerebellar atrophy in patients with
Teratog. Carcinog. Mutagen. 12 (1), 43–50. long term phenytoin exposure and epilepsy. Arch. Neurol. 51 (8), 767–771.
Kokenge, R., Kutt, H., McDowell, F., 1965. Neurological sequelae folowing dilantin Nolan, S.J., Marson, A.G., Pulman, J., Tudur Smith, C., 2013a. Phenytoin versus valproate
overdose in a patient and in experimental animals. Neurology 15, 823–829. monotherapy for partial onset seizures and generalised onset tonic-clonic seizures.
Komatsu, T., Yamazaki, H., Asahi, S., Gillam, E.M.J., Guengerich, F.P., Nakajima, M., Cochrane Database Syst. Rev. 8, CD001769.
Yokoi, T., 2000. Formation of a dihydroxy metabolite of phenytoin by human liver Nolan, S.J., Tudur Smith, C., Pulman, J., Marson, A.G., 2013b. Phenobarbitone versus
microsomes/cytosol: roles of cytochrome P450 2C9, 2C19, and 3A4. Drug Metab. phenytoin monotherapy for partial onset seizures and generalised onset tonic-clonic
Dispos. 28, 1361–1368. seizures. Cochrane Database Syst. Rev. 1, CD002217.
Kopsky, D.J., Keppel Hesselink, J.M., 2017. Topical phenytoin for the treatment of neu- Nseir, G., Golshayan, D., Barbey, F., 2013. Phenytoin-associated severe hypocalcemia
ropathic pain. J. Pain Res. 10, 469–473. with seizures in a patient with a TSC2-PKD1 contiguous gene syndrome. Ren. Fail. 35
Krishnamoorthy, K.S., Zalneraitis, E.L., Young, R.S., Bernad, P.G., 1983. Phenytoin-in- (6), 866–868.
duced choreoathetosis in infancy: case reports and a review. Pediatrics 72 (6), NTP (National Toxicology Program), 1993. Toxicology and carcinogenesis studies of 5,5-
831–834. diphenylhydantoin (CAS No. 57-41-0) (phenytoin) in F344/N rats and B6C3F1 mice
Kumar, N., Chakraborty, A., Suresh, S.H., Basappaji, S., Betdur, A.L., 2013. Phenytoin- (feed studies). Natl. Toxicol. Progr. Tech. Rep. 404, 1–303.
induced cerebellar atrophy in an epileptic boy. Indian J. Pharmacol. 45 (6), 636–637. O'Neil, M.J. (Ed.), 2001. The Merck Index - an Encyclopedia of Chemicals, Drugs, and
Kumkamthornkul, P., Udnaen, S., Tansit, T., Tuchinda, P., Srinoulprasert, Y., 2018. Biologicals, thirteenth ed. Merck and Co., Inc., Whitehouse Station, NJ, pp. 1312.
Evaluation of a lymphocyte transformation test and cytokine detection assay to Oelze, L.L., Pillow, M.T., 2013. Phenytoin-induced drug reaction with eosinophilia and

7
J. Patocka, et al. Food and Chemical Toxicology 142 (2020) 111393

systemic symptoms (DRESS) syndrome: a case report from the emergency depart- on anxiety, irritability, and anger in neurotic outpatients. Psychopharmacologia 17
ment. J. Emerg. Med. 44 (1), 75–78. (2), 169–181.
Okogbaa, J.I., Onor, I.O., Arije, O.A., Harris, M.B., Lillis, R.A., 2015. Phenytoin-induced Stille, G., Brunckow, I., 1954. [The ganglioplegic effect of 3-alkylaminoalkyl-hydantoins].
purple glove syndrome: a case report and review of the literature. Hosp. Pharm. 50 Arzneimittelforschung 4 (12), 723–725 (German).
(5), 391–395. Sugimoto, M., Wakabayashi, Y., Shiokawa, Y., Takaku, F., 1982. [Bone marrow heme
Onaolapo, A.Y., Adebayo, A.A., Onaolapo, O.J., 2018. Oral phenytoin protects against synthesis suppression by diphenylhydantoin in vitro in pure red cell aplasia]. Rinsho
experimental cyclophosphamide-chemotherapy induced hair loss. Pathophysiology Ketsueki 23 (9), 1446–1452 (Japanese).
25 (1), 31–39. Sullivan, J.R., Shear, N.H., 2001. The drug hypersensitivity syndrome: what is the pa-
Osorio, I., Reed, R.C., 1989. Treatment of refractory generalized tonic‐clonic status epi- thogenesis? Arch. Dermatol. 137 (3), 357–364.
lepticus with pentobarbital anesthesia after high‐dose phenytoin. Epilepsia 30 (4), SunshineI (Ed.), 1969. CRC Handbook of Analytical Toxicology. The Chemical Rubber
464–471. Co., Cleveland, pp. 44.
Ozkaynakci, A., Gulcebi, M.I., Ergeç, D., Ulucan, K., Uzan, M., Ozkara, C., Guney, I., Onat, Suwalsky, M., Mennickent, S., Norris, B., Cárdenas, H., 2006. The antiepileptic drug
F.Y., 2015. The effect of polymorphic metabolism enzymes on serum phenytoin level. phenytoin affects sodium transport in toad epithelium. Comp. Biochem. Physiol. C
Neurol. Sci. 36 (3), 397–401. Toxicol. Pharmacol. 142 (3–4), 253–261.
Prasad, M., Krishnan, P.R., Sequeira, R., Al-Roomi, K., 2014. Anticonvulsant therapy for Szabo, G.K., Pylilo, R.J., Davoudi, H., Browne, T.R., 1990. Simultaneous determination of
status epilepticus. Cochrane Database Syst. Rev. 9, CD003723. p-hydroxylated and dihydrodiol metabolites of phenytoin in urine by high-perfor-
Praveen-kumar, S., Desai, M., 2014. Ocular motor abnormalities in a patient with phe- mance liquid chromatography. J. Chromatogr., A 535 (1–2), 279–285.
nytoin toxicity–case report and minireview. Clin. Neurol. Neurosurg. 127, 116–117. Taing, K.D., O'Brien, T.J., Williams, D.A., French, C.R., 2017. Anti-epileptic drug com-
Raftopoulos, R., Hickman, S.J., Toosy, A., Sharrack, B., Mallik, S., Paling, D., Altmann, bination efficacy in an in vitro seizure model - phenytoin and valproate, lamotrigine
D.R., Yiannakas, M.C., Malladi, P., Sheridan, R., Sarrigiannis, P.G., Hoggard, N., and valproate. PloS One 12 (1), e0169974.
Koltzenburg, M., Gandini Wheeler-Kingshott, C.A., Schmierer, K., Giovannoni, G., Tan, E.K., Chan, L.L., Auchus, A.P., 2001. Phenytoin cerebellopathy without epilepsy.
Miller, D.H., Kapoor, R., 2016. Phenytoin for neuroprotection in patients with acute Acta Neurol. Scand. 104 (1), 61–62.
optic neuritis: a randomised, placebo-controlled, phase 2 trial. Lancet Neurol. 15 (3), Thangaraju, P., Tamilselvan, T., Venkatesan, S., Eswaran, T., Singh, H., Giri, V.C.,
259–269. Showkath Ali, M.K., 2015. Topical phenytoin for managing various ulcers:A meta-
Razmaraii, N., Babaei, H., Nayebi, A.M., Asadnasab, G., Helan, J.A., Azarmi, Y., 2016. analysis. Sudan Medical Monitor 10 (2), 63–67.
Cardioprotective effect of phenytoin on doxorubicin-induced cardiac toxicity in a rat Theil, G.B., Richter, R.W., Powell, M.R., Doolan, P.D., 1961. Acute dilantin poisoning.
model. J. Cardiovasc. Pharmacol. 67 (3), 237–245. Neurology 11, 138–142.
Reynolds, E.H., 1967. Schizophrenia-like psychoses of epilepsy and disturbances of folate Thomson, P.D.R., 2007. Micromedex. Drug Information for the Health Care Professional
and Vitamin B12 metabolism induced by anticonvulsant drugs. Br. J. Psychiatry 113 ume 1. Greenwood Village, CO, pp. 271 ISBN-13: 978-1563635144.
(501), 911–919. TOXNET Data reported in TOXNET. https://chem.nlm.nih.gov/chemidplus/rn/20830-
Riedel, L., Obe, G., 1984. Mutagenicity of antiepileptic drugs. II. Phenytoin, primidone 75-5 has been used. Original literature sources are unthinkable).
and phenobarbital. Mutat. Res. 138 (1), 71–74. Treiman, D.M., Woodbury, D.M., 1995. Phenytoin: absorption, distribution, and excre-
Sadeghi, L., Yekta, R., Dehghan, G., 2018. New mechanisms of phenytoin in calcium tion. In: Levy, R.H., Mattson, R.H., Meldrum, B.S. (Eds.), Antiepileptic Drugs, fourth
homeostasis: competitive inhibition of CD38 in hippocampal cells. Daru 26 (2), ed. Raven Press, New York, pp. 301–314.
191–198. Urashima, Y., Urashima, K., Ohnishi, M., Matsushita, K., Suzuki, K., Kurachi, K.,
Safari, J., Naeimi, H., Ghanbari, M.M., Fini, O.S., 2009. Preparation of phenytoin deri- Nishihara, M., Katsumata, T., Myotoku, M., Ikeda, K., Hirotani, Y., 2019. Interaction
vatives under solvent-free conditions using microwave irradiation. Russ. J. Org. between phenytoin and enteral nutrients and its influence on gastrointestinal ab-
Chem. 45 (3), 477–479. sorption. Pharmazie 74 (9), 559–562.
Salinsky, M.C., Oken, B.S., Binder, L.M., 1996. Assessment of drowsiness in epilepsy second ed. Usdin, E., Efron, D.H. (Eds.), 1972. Psychotropic Drugs and Related
patients receiving chronic antiepileptic drug therapy. Epilepsia 37 (2), 181–187. Compounds –. pp. 224 Washington DC.
Sangster, J., 1994. LOGKOW Databank. Sangster Res. Lab. Canada, Montreal Quebec. Velasco, M.J., McDermott, J., 2014. Drug rash with eosinophilia and systemic symptoms
Sasaki, H., Ikedo, D., Kataoka, M., Kido, J.I., Kitamura, S., Nagata, T., 1999. Pronounced (DRESS) syndrome and hepatitis induced by phenytoin. Int. J. Dermatol. 53 (4),
palatal and mandibular tori observed in patient with chronic phenytoin therapy: a 490–493.
case report. J. Periodontol. 70 (4), 445–448. Weichbrodt, G.D., Elliott, D.P., 1987. Treatment of phenytoin toxicity with repeated doses
Schwinghammer, T.L., Howrie, D.L., 1983. Phenytoin-induced lymphadenopathy. Drug of activated charcoal. Ann. Emerg. Med. 16 (12), 1387–1389.
Intell. Clin. Pharm. 17 (6), 460–462. Wilson, J.T., Höjer, B., Rane, A., 1976. Loading and conventional dose therapy with
Scott, J.M., Weir, D.G., 1980. Drug-induced megaloblastic change. Clin. Haematol. 9 (3), phenytoin in children: kinetic profile of parent drug and main metabolite in plasma.
587–606. Clin. Pharmacol. Ther. 20 (1), 48–58.
Scriba, G.K., Lambert, D.M., Poupaert, J.H., 1995. Bioavailability of phenytoin following Wu, X., Liu, W., Zhou, W., 2018. Association of CYP2C9*3 with phenytoin-induced
oral administration of phenytoin-lipid conjugates to rats. J. Pharm. Pharmacol. 47 Stevens-Johnson syndrome and toxic epidermal necrolysis: a systematic review and
(11), 945–948. meta-analysis. J. Clin. Pharm. Therapeut. 43 (3), 408–413.
Selhorst, J.B., Kaufman, B., Horwitz, S.J., 1972. Diphenylhyadantoin induced cerebellar Yabuki, S., Nakaya, K., 1976. Immunoglobulin abnormalities in epileptic patients treated
degeneration. Arch. Neurol. 27 (5), 453–455. with diphenylhydantoin. Folia Psychiatr. Neurol. Jpn. 30 (2), 93–109.
Sezzano, P., Raimondi, A., Arboix, M., Pantarotto, C., 1982. Mutagenicity of diphe- Yalkowsky, S.H., He, Y., 2003. Handbook of Aqueous Solubility Data: an Extensive
nylhydantoin and some of its metabolites towards salmonella typhimurium strains. Compilation of Aqueous Solubility Data for Organic Compounds Extracted from the
Mutat. Res. 103 (3–6), 219–228. AQUASOL dATAbASE. CRC Press LLC, Boca Raton, FL, pp. 998.
Shanmugarajah, P.D., Hoggard, N., Aeschlimann, D.P., Aeschlimann, P.C., Dennis, G.J., Yamazaki, H., Hiroki, S., Urano, T., Inoue, K., Shimada, T., 1996. Effects of roxi-
Howell, S.J., Reuber, M., Grünewald, R.A., Hadjivassiliou, M., 2018. Phenytoin-re- thromycin, erythromycin and troleandomycin on their N-demethylation by rat and
lated ataxia in patients with epilepsy: clinical and radiological characteristics. Seizure human cytochrome P450 enzymes. Xenobiotica 26, 1143–1153.
56, 26–30. Yampayon, K., Sukasem, C., Limwongse, C., Chinvarun, Y., Tempark, T.,
Silvado, C.E., Terra, V.C., Twardowschy, C.A., 2018. CYP2C9 polymorphisms in epilepsy: Rerkpattanapipat, T., Kijsanayotin, P., 2017. Influence of genetic and non-genetic
influence on phenytoin treatment. Pharmgenomics Pers Med 11, 51–58. factors on phenytoin-induced severe cutaneous adverse drug reactions. Eur. J. Clin.
Şimşek, G., Ciftci, O., Karadag, N., Karatas, E., Kizilay, A., 2014. Effects of topical phe- Pharmacol. 73 (7), 855–865.
nytoin on nasal wound healing after mechanical trauma: an experimental study. Yasumori, T., Chen, L.S., Li, Q.H., Ueda, M., Tsuzuki, T., Goldstein, J.A., Kato, R.,
Laryngoscope 124 (12), E449–E454. Yamazoe, Y., 1999. Human CYP2C-mediated stereoselective phenytoin hydroxylation
Singh, G., Driever, P.H., Sander, J.W., 2005. Cancer risk in people with epilepsy: the role in Japanese: difference in chiral preference of CYP2C9 and CYP2C19. Biochem.
of antiepileptic drugs. Brain 128 (Pt 1), 7–17. Pharmacol. 57 (11), 1297–1303.
Siniscalchi, A., Gallelli, L., Russo, E., De Sarro, G., 2013. A review on antiepileptic drugs- Zhang, Cai, Kalueff, Allan V., Song, Cai, 2019. Minocycline ameliorates anxiety-related
dependent fatigue: pathophysiological mechanisms and incidence. Eur. J. Pharmacol. self-grooming behaviors and alters hippocampal neuroinflammation, GABA and
718 (1–3), 10–16. serum cholesterol levels in female Sprague-Dawley rats subjected to chronic un-
Smythe, M.A., Umstead, G.S., 1989. Phenytoin hepatotoxicity: a review of the literature. predictable mild stress. Behav. Brain Res. 363, 109–117. https://doi.org/10.1016/j.
DICP 23 (1), 13–18. bbr.2019.01.045.
Somberg, J., 1985. Antiarrhythmic drug therapy. Recent advances and current status. Zhang, L., Li, Z., Ma, G., Han, X., Li, C., Shan, M., Chen, L.A., 2018. Systematic review of
Cardiology 72 (5–6), 329–348. phenytoin intoxication induced by compound phenytoin sodium, ephedrine hydro-
Srinivasan, G., Wyawahare, M., Mathen, P.G., Subrahmanyam, D.K., 2015. Life-threa- chloride and theophylline tablets in China. Medicine (Baltim.) 97 (51), e13689.
tening bradyarrhythmia with oral phenytoin overdose. J. Pharmacol. Pharmacother. Zinsmeister, S., Marks, R.E., 1976. Acute athetosis as a result of phenytoin toxicity in a
6 (3), 179–181. child. Am. J. Dis. Child. 130 (1), 75–76.
Stephens, J.H., Shaffer, J.W., 1970. A controlled study of the effects of diphenylhydantoin

You might also like