J FCT 2020 111393

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 46

Journal Pre-proof

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


toxicology

Jiri Patocka, Qinghua Wu, Eugenie Nepovimova, Kamil Kuca

PII: S0278-6915(20)30281-7
DOI: https://doi.org/10.1016/j.fct.2020.111393
Reference: FCT 111393

To appear in: Food and Chemical Toxicology

Received Date: 1 January 2020


Revised Date: 16 April 2020
Accepted Date: 24 April 2020

Please cite this article as: Patocka, J., Wu, Q., Nepovimova, E., Kuca, K., Phenytoin – An anti-seizure
drug: Overview of its chemistry, pharmacology and toxicology, Food and Chemical Toxicology (2020),
doi: https://doi.org/10.1016/j.fct.2020.111393.

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition
of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of
record. This version will undergo additional copyediting, typesetting and review before it is published
in its final form, but we are providing this version to give early visibility of the article. Please note that,
during the production process, errors may be discovered which could affect the content, and all legal
disclaimers that apply to the journal pertain.

© 2020 Published by Elsevier Ltd.


Phenytoin – an Anti-Seizure Drug: Overview of its Chemistry,

Pharmacology and Toxicology

Jiri Patocka,1,2 Qinghua Wu,3,4 Eugenie Nepovimova,3 and Kamil Kuca2,3*

1. Faculty of Health and Social Studies, Department of Radiology and Toxicology, University

of South Bohemia Ceske Budejovice, Ceske Budejovice, Czech Republic.

2. Biomedical Research Centre, University Hospital, Hradec Kralove, Czech Republic.

3. Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec

Kralove, Czech Republic.


4
College of Life Science, Yangtze University, Jingzhou 434025 China

Corresponding author: Prof. Kamil Kuca

kamil.kuca@uhk.cz

+420 603 289 166

ORCID: 0000-0001-9664-1109

Acknowledgement

This work was supported be Czech Science Foundation project: 17-19968-S.


Phenytoin – an Anti-Seizure Drug: Overview of its Chemistry,

Pharmacology and Toxicology

Abstract

Phenytoin is a long-standing, anti-seizure drug widely used in clinical practice. It has also

been evaluated in the context of many other illnesses in addition to its original epilepsy

indication. The narrow therapeutic index of phenytoin and its ubiquitous daily use pose a high

risk of poisoning. This review article focuses on the chemistry, pharmacokinetics, and

toxicology of phenytoin, with a special focus on its mutagenicity, carcinogenicity, and

teratogenicity. The side effects on human health associated with phenytoin use are thoroughly

described. In particular, DRESS syndrome and cerebellar atrophy are addressed. This review

will help in further understanding the benefits phenytoin use in the treatment of epilepsy.

Keywords: phenytoin; pharmacokinetics; toxicity; side effect; DRESS syndrome; cerebellar

atrophy.
INTRODUCTION

Phenytoin is an anti-seizure drug that has been under clinical evaluation for around eight

decades. It is primarily used for the treatment of tonic-clonic and partial seizures (Abou-

Khalil, 2016). Phenytoin has a highly selective inhibitory effect on the motor area of the

cerebral cortex. For its mode of action, phenytoin binds to the inactivated state of the Na+

channel to prolong the neuronal refractory period (Keppel Hesselink, 2017). Moreover, it is

generally believed that phenytoin exerts its antiepileptic effect by stabilising the function of

brain cell membranes and increasing the levels of the inhibitory neurotransmitters serotonin

(5-HT) and γ-aminobutyric acid (GABA) in the brain (Keppel Hesselink, 2017; Keppel

Hesselink and Kopsky, 2017a,b). Through these mechanisms, phenytoin prevents the spread

of abnormal discharge and has anti-epileptic effects. The anti-neuralgia activity of phenytoin

may also be related to these same mechanisms; a reduction in synaptic transmission or a

reduction in transient stimuli that cause neuronal discharge (Hall et al. 2020). In cardiac

tissue, phenytoin inhibits the ectopic rhythm of the atrium and ventricle, and accelerates the

conduction of the atrioventricular node to reduce myocardial autonomy, producing an

antiarrhythmic effect (Zhang et al. 2018).

Phenytoin can be administered by oral delivery or parenteral delivery. The intravenous

version of this drug is usually made available as a sodium salt, and this can be adapted to a

propylene glycol-alcohol-water solution for parenteral administration of phenytoin for

treatment of status epilepticus, especially where no further improvement is observed with

benzodiazepines (Prasad et al., 2014). Occasionally, neuropathic pain or heart arrhythmias are

also treated in this fashion (Somberg, 1985; Kopsky et al., 2017). In addition, neurological,

psychiatric, and non-CNS indications of phenytoin have been identified, such as wound
healing, and several of these indications have been investigated in subsequent pilot studies

(Şimşek et al., 2014). Wound healing is one of the indications most thoroughly investigated

(Keppel Hesselink, 2018; Barratt et al., 1997).

However, phenytoin treatment is associated with several side effects, and toxicity plays a

key role in these issues (Imam et al., 2014). An excessive dose of phenytoin inhibits the

central nervous system, causes cerebellar dysfunction, dyskinesia, and induces peripheral

neuropathy. In addition, phenytoin can cause gingival hyperplasia, induce pseudolymphomas

and malignant lymphomas, and cause allergic reactions (Keppel Hesselink, 2017; Farook et al.

2019). As mentioned above, intravenous injections of phenytoin slow the conduction rate

between myocardial fibres and eliminate ectopic rhythms, producing a similar effect to

quinidine (Mathews et al. 2019). In addition, phenytoin can cause cerebellar syndrome, which

manifests itself as ataxia, nystagmus, hand tremor, and diplopia. These manifestations of

cerebellar syndrome are dependent on phenytoin dose and blood concentration; tremor occurs

at an average plasma concentration of 20 µg/mL, and ataxia occurs at 30 µg/mL. A coma can

occurred at average plasma concentrations of 40 µg/mL or more (Zhang et al. 2018). In the

long-term, sudden seizures may become worse during phenytoin treatment, although seizures

can become more worse following withdrawal of this medicine.

Phenytoin is more and more widely used in clinical practice, including for wound healing,

migraine, dizziness, hiccups, myocardial infarction, and burns (Keppel Hesselink and Kopsky,

2017a,b). Therefore, the development and utilisation of this drug has attracted increasing

attention. However, the adverse reactions associated with phenytoin treatment should not be

ignored; known symptoms include gastrointestinal irritation, gum hyperplasia, allergic

reactions, and mental health issues (Sadeghi et al. 2018). To prevent such serious adverse

consequences, it is necessary that phenytoin is used reasonably in clinical practice. This


review article focuses on the chemistry, mechanism of action, pharmacokinetics, toxicology

of phenytoin, and its side effects on human health.

HISTORY

The German chemist Heinrich Biltz synthesised phenytoin (diphenylhydantoin) for the

very first time in 1908. He later sold his work to Parke-Davis (Keppel Hesselink and Kopsky,

2017). The additional usefulness of phenytoin was revealed in 1938 by Tracy Putnam and H.

Houston Merritt, when they discovered that it could be used to control seizures, while

avoiding the sedation that was typical to phenobarbital (Merritt and Putnam, 1984). Phenytoin

has been examined and evaluated in the context of many illnesses subsequent to its original

epilepsy indication. For several disorders, phenytoin is still undergoing some form of

innovation, including for bipolar disorder and wound healing, amongst others (Keppel

Hesselink, 2018). Using meta-analysis, it has also been revealed that phenytoin may be used

to manage different types of ulcers (Thangaraju et al., 2015). The use of phenytoin as a

neuroprotector, in the context of multiple sclerosis, has also been explored in clinical trials

(Raftopoulos et al., 2016).

CHEMISTRY

Phenytoin (Figure 1) is a hydantoin derivative (5,5-difenylhydantoine, 5,5'-

diphenylimidazolidine-2,4-dione; C15H12N2O2; M.W. 252.27; CAS Registry Number 57-41-

0). It is a fine white or almost white crystalline powder with the following characteristics:
m.p. 295 - 298°C, (O'Neil, 2001); pKa = 8.33, (Sangster, 1994); log P (octanol water) = 2.47,

(Hansch et al., 1995); slightly soluble in water (32 mg/L), (Yalkowsky and He, 2003).

Moreover, phenytoin is soluble in alkali hydroxides, alcohol, acetone (O'Neil, 2001) and

acetic acid (Lide and Milne, 1994). Phenytoin is sensitive to light (Sunshine, 1969) and

solutions are yellowed in light (Thompson and Micromedex, 2007). Finally, phenytoin is

odourless (or almost odourless) and tasteless (IARC, 1972).

There are two methods through which phenytoin can be synthesised. Phenytoin synthesis

can be achieved by adding base-catalysed urea to benzyl to produce benzilic acid, from which

phenytoin is produced following a rearrangement. This process is called Biltz synthesis of

phenytoin (Safari et al., 2009). In addition, phenytoin can be produced by oxidation of

benzoin with nitric acid to produce the benzyl, after which the procedure is the same as for

Biltz synthesis. In this process, almonds may be used as a source of benzaldehyde (Ashnagar

et al., 2009). Water-soluble phenytoin derivatives have also been synthesised (Bosch et al.,

1999). It should be noted that during decomposition caused by heat, phenytoin typically

produces several toxic fumes, including nitrogen oxide, carbon monoxide and carbon dioxide

(Lewis, 2004).

MECHANISM OF ACTION

The mechanism of action of phenytoin in neuropharmacology has been investigated for

more than 80 years (Keppel Hesselink, 2017). Phenytoin is a voltage-gated, sodium channel

blocker (Hains et al., 2004). It exerts its effect by stabilising the inactive state of the Na+

channel and prolonging the neuronal refractory period (Keppel Hesselink, 2017; Keppel
Hesselink and Kopsky, 2017a,b). Through this mode of action, phenytoin regulates the

bioelectrical activities of various systems in humans. Importantly, phenytoin inhibits or

eliminates abnormal electrical activity in nerve and muscle cells without affecting the

production and conduction of normal bioelectricity. The electrophysiological basis of this

effect involves the regulation of the transmembrane movement and intracellular distribution

of Na+, K+, and Ca2+ (Keppel Hesselink, 2017; Martin et al. 2014). Phenytoin has an

especially significant blocking effect on the Na+ channels of neurons with high-frequency

abnormal discharge, where it inhibits these repeated high-frequency discharges; in contrast,

phenytoin has no significant effect on normal low-frequency discharges (Martin et al. 2014).

In addition, phenytoin inhibits the rapidly inactivating (T-type) Ca2+ channels of neurons and

inhibits Ca2+ influx (Keppel Hesselink and Kopsky, 2017a). Through this mechanism,

phenytoin affects the transmission of Ca2+-dependent multiple neurotransmitters at the

synapse, and the post-synaptic responses.

Phenytoin also regulates hormone secretion of endocrine glands cells and stimulates

metabolism in the liver drug enzyme system. Moreover, phenytoin increases high density

lipoprotein concentration, and stimulates granulation and capillary formation in healing

tissues (Taing et al. 2017; Keppel Hesselink, 2017). The stability of phenytoin effects on cell

membrane bioelectric activity is manifested in its ability to correct excessive cell excitement

caused by various activities: phenytoin can correct the excessive excitability of squid giant

axonal cells caused by high-frequency electrical stimulation or low Ca2+ and Mg2+; phenytoin

can increase resting potential in a high-potassium depolarisation state, and reduce the

frequency of endplate electrical sobbing, thereby affecting the motor nerve endplate potential

(Suwalsky et al. 2006). In a recent study, phenytoin was shown to inhibit Ca2+ in CD38

pathways and cause oxidative stress, inflammation, and depression (Sadeghi et al. 2018).
Phenytoin regulates a series of neurotransmitters including acetylcholine, serotonin,

norepinephrine, dopamine, GABA, and endorphins (Chou et al. 2014; Granger et al. 1995).

At high concentrations, phenytoin can inhibit GABA uptake at nerve endings, indirectly

enhancing the role of GABA by inducing GABA receptor proliferation; the end result is an

increase in Cl- influx and hyperpolarisation, and subsequent inhibition of the incidence and

spread of abnormal high-frequency discharge (Chou et al. 2014; Zhang et al. 2019). While

phenytoin 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 stimulating 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 activity, 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

prevented and activity in the brain that is known to cause the tonic-clonic seizure’s tonic

phase is diminished (Osorio and Reed, 1989).

PHARMACOKINETICS

Absorption, distribution, metabolism, and excretion (ADME)

Phenytoin is well absorbed following oral administration (Urashima et al. 2019). Its

absorption takes place predominantly in the duodenum. The rate of dissolution of phenytoin

in the intestines informs its level of absorption. Peak plasma concentrations are typically

reached during a four to eight-hour period. The distribution of the drug is widespread
throughout the body (distribution volume 0.8 L/kg). Phenytoin easily crosses the blood –

brain barrier and it is extensively bound (∼90%) to plasma albumin (Treiman and Woodbury,

1995). Phenytoin also easily crosses the placenta (Mirkin, 1971). Indeed, similar plasma

concentrations of phenytoin have been measured in maternal and umbilical cord (Nau et al.,

1982). However, levels are typically low in breast milk obtained from mothers with epilepsy

(Mirkin, 1971).

Phenytoin is metabolised by cytochrome P450 enzyme to 5-(p-hydroxyphenyl)-5-

phenylhydantoin (4'-HPPH). Further metabolisation to a catechol is possible, and this can

extemporaneously oxidise to quinone and semiquinone species (Cuttle et al., 2000;

Ozkaynakci et al. 2015). Phenytoin metabolism has been widely studied in vitro with respect

to human subjects (Yasumori et al., 1999; Komatsu et al., 2000) and in vivo (Maguire, 1988;

Szabo et al., 1990). Four oxidative metabolites are reported, 4′-HPPH, 3′-HPPH, 3′,4′-

diHPPH, and 3′,4′-dihydrodiol (Maguire, 1988; Szabo et al., 1990). Research suggests that

phenytoin is oxidised to 4′-HPPH by CYP2C9 in humans, and to a minor extent by CYP2C19

(Bajpai et al., 1996). However, the role played by the P450s in the development of other

metabolites requires further investigation. Recent reports suggest that the contribution of

CYP3A4, CYP2C9, and CYP2C19 to liver microsomal 3′,4′-diHPPH formation from primary

hydroxylated metabolites differs according to the individual person (Komatsu et al., 2000;

Lakehal et al. 2002). CYP2C9 variants are observed to play a role in the metabolism of

phenytoin in the Chinese population (Chen et al. 2016). Furthermore, according to recent

studies, phenytoin has much higher CYP3A4-inducing potency. Therefore, CYP3A4-

metabolised drugs may generally be required during concurrent treatment with phenytoin

(Hole et al. 2018).

Research has also been conducted to study phenytoin metabolism in animal subjects

(Chow et al., 1980; Billings, 1983; Doecke et al., 1990). It should be noted that rates of 4′-
HPPH formation in rat liver microsomes were 10 times higher than in humans (Munns et al.,

1997). The reasons for this difference in P450 metabolism are still unclear. Phenytoin has

been shown to induce several forms of P450 (Fleishaker et al., 1995; Ghosal et al., 1996;

Yamazaki et al., 1996). However, it is unknown whether administrating phenytoin can lead to

auto-induced metabolism in humans and rats (Edeki and Brase, 1995; Chetty et al., 1998).

Phenytoin displays non-linear elimination pharmacokinetics (Browne & LeDuc, 1995). A

result of this non-linearity is that the elimination half-life differs with plasma concentration. A

saturation of metabolite-inducing enzymes is the reason for the non-linearity in the

elimination of phenytoin (Browne & LeDuc, 1995). As shown in Figure 2, the main primary

metabolites are 5(3'-hydroxyphenyl)-5-phenylhydantoin (3′-HPPH), 5(4'-hydroxyphenyl)-5-

phenylhydantoin (4′-HPPH), 5(3',4'-dihydroxyphenyl)-5-phenylhydantoin (3′,4′-diHPPH),

and 5(3',4'-dihydroxy-1',5'-cyclohexydiene-1-yl)-5-phenylhydantoin (3′,4′-dihydrodiol). In

addition, arene-oxide is a highly reactive and extremely unstable hypothetical metabolite

which has never been isolated. Arene oxide may be responsible for the carcinogenic effects of

phenytoin. The main secondary metabolites are glucuronide conjugates of phenytoin and its

hydroxylated derivatives (Scriba et al. 1995).

SIDE EFFECTS OF PHENYTOIN

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

(Hwang and Tsai, 2004). These include drowsiness (Salinsky et al., 1996), fatigue

(Siniscalchi et al., 2013), loss of control of bodily movements and loss of balance or

coordination (Iivanainen and Savolinen, 1983), irritability (Stephens and Shaffer, 1970),

restlessness (Drake, 1988), and several involuntary functions (Ahmad et al., 1975; Gunduz et
al., 2013) including involuntary eye movements (Bittencourt et al., 1980; Matsue et al.,

1981).

Phenytoin can cause cutaneous adverse reactions, from maculopapular exanthema to

SCAR (severe cutaneous adverse reaction), including drug reactions with eosinophilia, and

systemic symptoms, including Stevens-Johnson syndrome and toxic epidermal necrolysis.

The pharmacogenomic basis of these severe cutaneous adverse side effects can be linked to

genetic factors (Chung et al., 2014).

Typically, the reactions include poor coordination, an increase in the growth of hair,

stomach aches, loss of appetite, nausea, and gingival enlargement (Arya et al., 2012; Chacko

and Abraham, 2014; Onaolapo et al., 2018). Potentially more serious side effects include liver

problems (Lee et al., 1976; Curry et al., 2018), bone marrow suppression (Sugimoto et al.,

1982) and toxic epidermal necrolysis (Wu et al., 2018). Hepatotoxicity related to phenytoin is

a serious idiosyncratic side effect that is observed in a small percentage of those undergoing

treatment. It is accompanied by a host of symptoms ranging from a fever to arthralgias

(Smythe and Umstead, 1989). In some patients, phenytoin can cause liver necrosis (Lee et al.,

1976).

Many other syndromes have been attributed to phenytoin use. These include: paradoxical

seizure (Chua et al., 1999); drug withdrawal seizure (Nolan et al., 2013a,b); iplopia, blurred

vision, and colour disturbances (López et al., 1999); encephalopathy (Mehndiratta, 2016);

ventricular fibrillation (Merlo González et al., 2002); atrioventricular conduction disorder

(Magadle et al., 1999); enlarged lymph nodes (Schwinghammer and Howrie, 1983); purple

spots on the skin (Oelze and Pillow, 2013); and maculopapular exanthema leading to severe

cutaneous adverse side effects. These are supplemental to the common side effects described

earlier, such as systematic and eosinophilia symptoms, toxic epidermal necrolysis, Stevens-
Johnson syndrome (Chung et al., 2014), coarsening of facial features (Sasaki et al., 1999),

periarteritis nodosa (Haas, 1967), megaloblastic (folate-deficiency) anaemia (Scott and Weir,

1980), low blood calcium levels (hypocalcaemia) (Nseir et al., 2013), and immunoglobulin

abnormalities (Yabuki and Nakaya, 1976).

In addition, adverse effects related with phenytoin use can include various psychiatric

disorders or behavioural symptoms, specifically those associated with vitamin B12 or a folic

acid deficiency (Reynolds, 1967; Matsuura, 1999; Gatzonis et al., 2003). A rare case of

toxicity was recently reported that presented itself as psychosis resulting from vitamin

deficiency (Borasi et al., 2015). However, it is necessary to recognise that psychological and

behavioural side effects occur more frequently in patients taking antiepileptics other than

phenytoin (Chen et al., 2017).

DRESS syndrome

Allergic reactions to phenytoin present themselves as a rash. They can sometimes, if rarely,

take up more severe forms, e.g. anaphylaxis or DRESS (drug reaction with eosinophilia and

systemic symptoms) (Kumkamthornkul et al. 2018). A side effect linked to DRESS syndrome

is the drug-induced hypersensitivity syndrome (amongst other names). This side effect is a

known SCAR (severe cutaneous adverse reaction) and can prove to be life-threatening

(Sullivan and Shear, 2001). Drug-induced hypersensitivity syndrome is associated with

phenytoin in most cases and was originally referred to as phenytoin hypersensitivity

syndrome (De et al., 2018). Although this syndrome was subsequently discovered to occur

with several different drugs, phenytoin remains a frequent cause of this allergic reaction

(Brizendine and Naik, 2013; Deka et al., 2013; Hall and Fromm, 2013; Velasco and

McDermott, 2014; Yampayon et al., 2017). DRESS typically has a two to six-week latency

period. While it is known that the pathophysiology of DRESS involves the activation of
lymphocytes and reactivation of viruses, its pathophysiology is not completely understood

(Yampayon et al., 2017). DRESS is normally associated with a morbilliform cutaneous

eruption, and this is accompanied by lymphadenopathy, and a rise in temperature. The

severity of DRESS is linked to systemic involvement, and it can result in multiple organ-

failure (Ghannam et al., 2017).

Cerebellar atrophy

The adverse side effects of phenytoin can be chronic and/or acute. As the amount of

phenytoin rises, there is a disproportionate rise in the plasma drug concentration, despite the

increments in dosage itself being small, and this increase in plasma drug concentration is

accompanied by a change from subtherapeutic to toxic levels (McNamara, 1996). These

plasma drug concentration changes are highly linked to CNS dysfunction, viz. ataxia,

drowsiness, and nystagmus (Browne, 1997). However, these reactions can be reversed when

the dosage is reduced. A chronic overdose has been found to be responsible for cerebellar

atrophy in adult patients being administered phenytoin (Kokenge et al., 1965; Selhorst et al.,

1972; Ghatak et al., 1976; Mc Lain et al., 1980; Baier et al., 1984; Ney et al., 1994; Ahuja et

al., 2000; Tan et al., 2001; De Marcos et al., 2003; Lee et al., 2003; Chow and Szeto, 2007;

Kumar et al., 2013). Evidence has been reported that CYP2C9 polymorphisms are associated

with an increase in the frequency of cerebellar atrophy following the use of phenytoin

(Silvado et al., 2018).


TOXICOLOGY

The therapeutic range for phenytoin is narrow and a total serum concentration greater than

80 µM is, for many patients, linked to clinically relevant toxicity (Praveen-kumar and Desai,

2014). Phenytoin is typically excreted by the kidneys after it has been metabolised by hepatic

enzymes. However, a toxic accumulation of phenytoin can present itself in the context of

people with renal failure (Chua et al., 2000). Haemodialysis does not remove phenytoin

because it is 90% albumin bound (Martin et al., 1977). While this form of toxicity is not

typically fatal, it can lead to neurological symptoms ranging from a coma (Craig, 2005), to

nystagmus (Praveen-kumar and Desai, 2014), to ataxia (Shanmugarajah et al., 2018). On rare

occasions, intravenous applications may face complications stemming from Purple Glove

Syndrome (PGS) (Garbovsky et al., 2015). PGS is a serious side effect that can lead to

amputation (Jaain et al., 2015; Okogbaa et al., 2015).

Toxicity relating to phenytoin can be a product of dose misuse, dose adjustment, an

interaction with other drugs, or a patient’s changing physiology (Kang et al., 2013; Srinivasan

et al., 2015). The first symptoms of phenytoin intoxication are nausea, dysfunctions related to

the central nervous system (such as ataxia or nystagmus), a depressed state, and coma (Craig,

2005). Problems such as hypotension or arrhythmia are not typically linked to the use of this

drug. However, in cases of parenteral administration, these problems may present themselves.

Phenytoin is considered a moderately toxic substance (Imam et al., 1990). On its own, the

likelihood that phenytoin intoxication will result in patient death is considered to be very low.

The published acute toxicity data for phenytoin in different organisms are summarised in

Table 1. Data on mutagenicity, carcinogenicity, and teratogenicity are provided in separate

subchapters.
The mutagenicity of phenytoin and the mutagenicity of its major metabolite, 5-(4-

hydroxyphenyl)-5-phenylhydantoin (HPPH), have been tested in vitro on different Salmonella

typhimurium strains (TA1535, TA100, TA1537, TA1538, TA98) using an Ames test (Sezzano

et al., 1982; Léonard et al., 1984; Riedel and Obe, 1984). From the results of these studies, it

was concluded that phenytoin does not have mutagenic properties.

The experimental approach used in earlier mutagenicity tests (Montes de Oca-Luna et al.,

1984; Barcellona et al., 1987) is likely to be responsible for generating reactive arene-oxide

(Daly et al., 1972), an extremely unstable metabolite that has never been isolated (Brown and

LeDuc, 1995). Therefore, phenytoin was found not to be directly genotoxic in the context of a

set of shorter-term assays using cytogenetics (Kindig et al., 1992).

Experiments in laboratory animals have not provided any evidence of phenytoin

carcinogenicity (Jang et al., 1987; Maeda et al., 19988; Chhabra et al., 1993); although in one

case the results are inconsistent (NTP, 1993). Hepatocellular neoplasia in phenytoin-treated

rodents have little to no relevance for humans. At present, in the context of epilepsy patients,

no link has been found between cancer of the liver and phenytoin (Dethloff et al., 1996; Singh

et al., 2005; Friedman et al., 2009).

There are some indications that phenytoin is linked to a larger risk of congenital

malformations (Danielsson et al., 2005; Eroğlu et al., 2008). The risk of congenital

malformations is linked to phenytoin’s antiepileptic activity. A good number of studies have

revealed that this is the main risk factor for a rise in the incidence of congenital

malformations. The risks associated with teratogenicity are multifactorial and include factors

such as a patient’s genetic predisposition (Canger et al, 1999).

Typically, therapy against phenytoin toxicity would involve supportive care. Therapy is

focussed on the vital functions of the patient’s body, alongside managing vomit and nausea.
In addition, supportive care is essential to prevent injuries that can otherwise result from

ataxia or confusion. There is no antidote to phenytoin toxicity, and little evidence to suggest

that enhanced elimination or gastrointestinal decontamination can speed up recovery (Zhang

et al. 2019).

CONCLUSIONS

Phenytoin is a long-standing antiepileptic drug which exerts its effects by reducing the

influx of Na+ and Ca2+ into nerve cells and promoting the release of GABA; thus phenytoin

effectively inhibits the abnormal discharge of neurons, prevents the transmission of abnormal

impulses, and reduces the symptoms of withdrawal. In the brain, opioid µ and δ receptors

mediate epilepsy-like reactions, and endogenous opioid peptides and opioid receptors are

involved in the pathophysiology of primary epilepsy. A correlation may exist between the

pathogenesis of addictive diseases and epilepsy. Therefore, antiepileptic drugs may be used to

treat addictive diseases. Phenytoin treatment can control withdrawal symptoms in heroin

addicts and can effectively reduce the use of alternatives, such as methadone, in the treatment

of opioid addicts without affecting their efficacy. Moreover, phenytoin effectively improves

symptoms of anxiety, craving, and protracted withdrawal.

Phenytoin-induced toxicity can occur following an increase in the daily dose, a change in

formulation or brand, or a change in the frequency of administration. In recent years, there

have been many reports of adverse reactions caused by therapeutic doses of phenytoin.

However, there is no specific antidote to phenytoin toxicity. Therefore, individualised

administration is of great significance for the safe and effective use of phenytoin. With the
widespread application of new molecular biology techniques, current research on

individualised phenytoin medication has gradually evolved from traditional drug therapy

monitoring to pharmacogenomics. From population pharmacokinetics, nonlinear mixed effect

modelling (NONMEM) has led to the development of a useful tool for designing gene-

oriented and individualised drug regimens.

In summary, phenytoin has a wide range of biological activities and pharmacological

indications. In addition to being used clinically to treat epilepsy, phenytoin is also used to

treat a variety of diseases, including migraine, dizziness, hiccups, myocardial infarction,

burns, and in promoting wound healing. Although phenytoin has been used in epilepsy

treatment for many years and has been somewhat superseded by newer anti-epileptic

medicines, there is good reason to believe that phenytoin will find new life as a drug for the

individualised treatment of epilepsy, addiction, migraine, and other diseases.

REFERENCES

Abou-Khalil BW. Antiepileptic Drugs. Continuum (Minneap Minn). 2016; 22: 132-156.

Ahmad S, Laidlaw J, Houghton GW, Richens A. Involuntary movements caused by phenytoin

intoxication in epileptic patients. J Neurol Neurosurg Psychiatry. 1975; 38(3): 225-231.

Ahuja SR, Karande S, Kulkarni MV. Cerebellar atrophy in an epileptic child: is it due to

phenytoin? J Postgrad Med. 2000; 46(4): 278-279.

Arya R, Gulati S. Phenytoin-induced gingival overgrowth. Acta Neurol Scand. 2012; 125(3):

149-155.
Ashnagar A, Gharib Naseri N, Amini M. Synthesis of 5,5-diphenyl-2,4-imisdazolidinedione

(Phenytoin) from Almond. Asian J Chem. 2009; 21(7): 4976-4980.

Baier WK, Beck U, Doose H, Klinge H, Hirsch W. Cerebellar atrophy following

diphenylhydantoin intoxication. Neuropediatrics 1984; 15(2): 76-81.

Bajpai M, Roskos LK, Shen DD, Levy RH. Roles of cytochrome P4502C9 and cytochrome

P4502C19 in the stereoselective metabolism of phenytoin to its major metabolite. Drug Metab

Dispos. 1996; 24(12): 1401-1403.

Barcellona PS, Barale R, Campana A, Zucconi D, Rossi V, Caranti S. Correlations between

embryotoxic and genotoxic effects of phenytoin in mice. Teratog Carcinog Mutagen. 1987;

7(2): 159-168.

Barratt ES, Stanford MS, Felthous AR, Kent TA. The effects of phenytoin on impulsive and

premeditated aggression: a controlled study. J Clin Psychopharmacol. 1997; 17(5): 341-349.

Billings RE. Sex differences in rats in the metabolism of phenytoin to 5-(3,4-

dihydroxyphenyl)-5-phenylhydantoin. J Pharmacol Exp Ther 1983; 225: 630–636.

Bittencourt PR, Gresty MA, Richens A. Quantitative assessment of smooth-pursuit eye

movements in healthy and epileptic subjects. J Neurol Neurosurg Psychiatry. 1980; 43(12):

1119-1124.

Borasi M, Verma RP, Gupta SK. Psychosis as Harbinger of Phenytoin Toxicity. Toxicol Int.

2015; 22(1): 160-161.

Bosch J, Roca T, Domènech J, Suriol M. Synthesis of water-soluble phenytoin prodrugs.

Bioorg Med Chem Lett. 1999; 9(13): 1859-1862.


Brizendine CE, Naik PJ. Drug rash with eosinophilia and systemic symptoms syndrome

associated with use of phenytoin, divalproex sodium, and phenobarbital. Am J Health Syst

Pharm. 2013; 70(6): 507-510.

Browne TR, LeDuc B. Phenytoin: chemistry and biotransformation. In: Levy RH, Mattson

RH, Meldrum BS, eds, Antiepileptic Drugs, 4th Ed., New York, Raven Press, pp. 1995; 283–

300.

Browne TR. Phenytoin and other hyadantoins. In: Engel J Jr, Pidley T, editors. Epilepsy. A

Comprehensive textbook. 1st Ed. Philadelphia: Lippincott-Raven; 1997. pp 1557-1579.

Canger R, Battino D, Canevini MP, Fumarola C, Guidolin L, Vignoli A, Mamoli D, Palmieri

C, Molteni F, Granata T, Hassibi P, Zamperini P, Pardi G, Avanzini G. Malformations in

offspring of women with epilepsy: a prospective study. Epilepsia. 1999; 40(9): 1231-1236.

Craig S. Phenytoin poisoning. Neurocrit Care. 2005; 3(2): 161-170.

Curry B, Mican L, Smith TL. Phenytoin-induced chronic liver enzyme elevation and hepatic

fibrosis: A case report. Ment Health Clin. 2018; 8(4): 184-187.

Cuttle L, Munns AJ, Hogg NA, Scott JR, Hooper WD, Dickinson RG, Gillam EM. Phenytoin

metabolism by human cytochrome P450: involvement of P450 3A and 2C forms in secondary

metabolism and drug-protein adduct formation. Drug Metab Dispos. 2000; 28(8): 945-950.

Daly JW, Jerina DM, Witkop B. Arene oxides and the NIH shift: the metabolism, toxicity and

carcinogenicity of aromatic compounds. Experientia. 1972; 28(10): 1129-1149.

Danielsson BR, Johansson A, Danielsson C, Azarbayjani F, Blomgren B, Sköld AC.

Phenytoin teratogenicity: hypoxia marker and effects on embryonic heart rhythm suggest an

hERG-related mechanism. Birth Defects Res Clin Mol Teratol. 2005; 73(3): 146-153.
De A, Rajagopalan M, Sarda A, Das S, Biswas P. Drug Reaction with Eosinophilia and

Systemic Symptoms: An Update and Review of Recent Literature. Indian J Dermatol. 2018;

63(1): 30-40.

De Marcos FA, Ghizoni E, Kobayashi E, Li LM, Cendes F. Cerebellar volume and long-term

use of phenytoin. Seizure. 2003; 12(5): 312-315.

Deka NM, Dass R, Das BK, Hoque R. Phenytoin induced DRESS syndrome. Indian J Pediatr.

2013; 80(3): 266.

Dethloff LA, Graziano MJ, Goldenthal E, Gough A, de la Iglesia FA. Perspective on the

carcinogenic potential of phenytoin based on rodent tumor bioassays and human

epidemiological data. Hum Exp Toxicol. 1996; 15(4): 335-348.

Doecke CJ, Sansom LN, McManus ME. Phenytoin 4-hydroxylation by rabbit liver P450IIC3

and identification of orthologs in human liver microsomes. Biochem Biophys Res Commun

1990; 166: 860–866.

Drake ME. Restless legs with antiepileptic drug therapy. Clin Neurol Neurosurg. 1988; 90(2):

151-154.

Edeki TI, Brase DA. Phenytoin disposition and toxicity: Role of pharmacogenetic and

interethnic factors. Drug Metab Rev 1995; 27: 449–469.

Eroğlu E, Gökçil Z, Bek S, Ulaş UH, Odabaşi Z. Pregnancy and teratogenicity of antiepileptic

drugs. Acta Neurol Belg. 2008; 108(2): 53-57.

Farook, FF; Nizam, MNM; Alshammari, A. An update on the mechanisms of phenytoin

induced gingival overgrowth. Open Dentistry J, 2019, 13: 430-435.


Fleishaker JC, Pearson LK, Peters GR. Phenytoin causes a rapid increase in 6β-

hydroxycortisol urinary excretion in humans—A putative measure of CYP3A induction. J

Pharm Sci 1995; 84: 292–294.

Friedman GD, Jiang SF, Udaltsova N, Quesenberry CP Jr, Chan J, Habel LA. Epidemiologic

evaluation of pharmaceuticals with limited evidence of carcinogenicity. Int J Cancer. 2009;

125(9): 2173-2178.

Gams RA, Neal JA, Conrad FG. Hydantoin-induced pseudo-pseudolymphoma. Ann Intern

Med. 1968; 69(3): 557-568.

Garbovsky LA, Drumheller BC, Perrone J. Purple Glove Syndrome after Phenytoin or

Fosphenytoin Administration: Review of Reported Cases and Recommendations for

Prevention. J Med Toxicol. 2015; 11(4): 445-459.

Gatzonis SD, Angelopoulos E, Sarigiannis P, Mantouvalos V, Ploumbidis D, Siafakas A.

Acute psychosis due to treatment with phenytoin in a nonepileptic patient. Epilepsy Behav.

2003; 4(6): 771–772.

Ghannam M, Mansour S, Nabulsi A, Abdoh Q. Anticonvulsant hypersensitivity syndrome

after phenytoin administration in an adolescent patient: a case report and review of literature.

Clin Mol Allergy. 2017; 15: 14.

Ghatak NR, Santoso RA, McKinney WM. Cerebellar degeneration following long term

phenytoin therapy. Neurology 1976; 26(9): 818-820.

Ghosal A, Sadrieh N, Reik L, Levin W, Thomas PE. (1996) Induction of the male-specific

cytochrome P450 3A2 in female rats by phenytoin. Arch Biochem Biophys 1996; 332: 153–

162.
Granger P, Biton B, Faure C (1995) Modulation of the gammaaminobutyric acid type A

receptor by the antiepileptic drugs carbamazepine and phenytoin. Mol Pharmacol 47(6):1189–

1196.

Graziani G, Barbadoro E, Testa R. Denzimol, a new anticonvulsant drug. III. Toxicological

evaluation. Arzneimittelforschung. 1983; 33(8): 1168-1173.

Gunduz T, Kocasoy-Orhan E, Hanagasi HA. Orolingual dyskinesia and involuntary neck

movements caused by phenytoin intoxication. J Neuropsychiatry Clin Neurosci. 2013; 25(4):

E51.

Haas P. [On a very acutely developing case of periarteritis nodosa following

diphenylhydantoin therapy]. Wien Klin Wochenschr. 1967; 79(3): 56-58. German.

Hains BC, Saab CY, Lo AC, Waxman SG. Sodium channel blockade with phenytoin protects

spinal cord axons, enhances axonal conduction, and improves functional motor recovery after

contusion SCI. Exp Neurol, 2004; 188(2): 365-377.

Hall DJ, Fromm JS. Drug reaction with eosinophilia and systemic symptoms syndrome in a

patient taking phenytoin and levetiracetam: a case report. J Med Case Rep. 2013; 7: 2.

Hall EA, Wheless JW, Phelps SJ. Status Epilepticus: The Slow and Agonizing Death of

Phenytoin. J Pediatr Pharmacol Ther. 2020, 25(1):4-6

Hansch C, Leo A, Hoekman D. Exploring QSAR - Hydrophobic, Electronic, and Steric

Constants. Washington, DC: American Chemical Society., 1995., p. 129.

Hesselink JMK. Phenytoin: a step by step insight into its multiple mechanisms of action—80

years of mechanistic studies in neuropharmacology. J Neurol. 2017; 264(9): 2043-2047.


Hole K, Wollmann BM, Nguyen C, Haslemo T, Molden E. Comparison of CYP3A4-inducing

capacity of enzyme-inducing antiepileptic drugs using 4β-hydroxycholesterol as biomarker.

Ther Drug Monit. 2018, 40(4):463-468.

Howrie DL, Crumrine PK. Phenytoin-induced movement disorder associated with intravenous

administration for status epilepticus. Clin Pediatr (Phila). 1985; 24(8): 467-469.

Hwang WJ, Tsai JJ. Acute phenytoin intoxication: causes, symptoms, misdiagnoses, and

outcomes. Kaohsiung J Med Sci. 2004; 20(12): 580-585.

Chacko LN, Abraham S. Phenytoin-induced gingival enlargement. BMJ Case Rep. 2014;

2014: pii: bcr2014204670.

Chao TI, Alzheimer C. Effects of phenytoin on the persistent Na+ current of mammalian CNS

neurones. Neuroreport, 1995; 6(13): 1778-1780.

Chen B, Choi H, Hirsch LJ, Katz A, Legge A, Buchsbaum R, Detyniecki K. Psychiatric and

behavioral side effects of antiepileptic drugs in adults with epilepsy. Epilepsy Behav. 2017;

76: 24-31.

Chen LG, Wang Z, Zhu Y, Xiong JH, Sun LR, Dai DP, Cai JP, Hu GX. In vitro metabolism

of phenytoin in 36 CYP2C9 variants found in the Chinese population. Chem Biol Interact.

2016, 253:93-99.

Chetty M, Miller R, Seymour MA. Phenytoin auto-induction. Ther Drug Monit 1998; 20: 60–

62.

Chhabra RS, Bucher JR, Haseman JK, Elwell MR, Kurtz PJ, Carlton BD. Comparative

carcinogenicity of 5,5-diphenylhydantoin with or without perinatal exposure in rats and mice.


Fundam Appl Toxicol. 1993; 21(2): 174-186. Erratum in: Fundam Appl Toxicol 1994; 22(1):

159.

Chou MY, Lee CY, Liou HH, Pan CY. Phenytoin attenuates the hyper-exciting

neurotransmission in cultured embryonic cortical neurons. Neuropharmacology. 2014, 83:54-

61.

Chow KM, Szeto CC. Cerebral atrophy and skull thickening due to chronic phenytoin

therapy. CMAJ. 2007; 176(3): 321-323.

Chow SA, Charkowski DM, Fischer LJ. Separation of phenytoin and its metabolites by high-

performance liquid chromatography. Life Sci. 1980; 27(25-26): 2477-2482.

Chua HC, Venketasubramanian N, Tan CB, Tjia H. Paradoxical seizures in phenytoin

toxicity. Singapore Med J. 1999; 40(4): 276-277.

Chua HC, Venketasubramanian N, Tjia H, Chan SP. Elimination of phenytoin in toxic

overdose. Clin Neurol Neurosurg 2000; 102(1): 6–8.

Chung WH, Chang WC, Lee YS, Wu YY, Yang CH, Ho HC, Chen MJ, Lin JY, Hui RC, Ho

JC, Wu WM, Chen TJ, Wu T, Wu YR, Hsih MS, Tu PH, Chang CN, Hsu CN, Wu TL, Choon

SE, Hsu CK, Chen DY, Liu CS, Lin CY, Kaniwa N, Saito Y, Takahashi Y, Nakamura R,

Azukizawa H, Shi Y, Wang TH, Chuang SS, Tsai SF, Chang CJ, Chang YS, Hung SI; Taiwan

Severe Cutaneous Adverse Reaction Consortium; Japan Pharmacogenomics Data Science

Consortium. Genetic variants associated with phenytoin-related severe cutaneous adverse

reactions. JAMA. 2014; 312(5): 525-534.

IARC. Monographs on the Evaluation of the Carcinogenic Risk of Chemicals to Humans.

Geneva: World Health Organization, International Agency for Research on Cancer, 1972.

Available at: http://monographs.iarc.fr/ENG/Classification/index.php, p. V66: 175.


Iivanainen M, Savolainen H. Side effects of phenobarbital and phenytoin during long-term

treatment of epilepsy. Acta Neurol Scand Suppl. 1983; 97: 49-67.

Imam SH, Landry K, Kaul V, Gambhir H, John D, Kloss B. Free phenytoin toxicity. Am J

Emerg Med. 2014; 32(10): 1301.e3-4.

Jain RS, Nagpal K, Kumar S, Prakash S, Handa R. Purple glove syndrome occurring after oral

administration of phenytoin in therapeutic doses: mechanism still a dilemma. Am J Emerg

Med. 2015; 33(1): 123.e5-6.

Jang JJ, Takahashi M, Furukawa F, Toyoda K, Hasegawa R, Sato H, Hayashi Y. Long-term in

vivo carcinogenicity study of phenytoin (5,5-diphenylhydantoin) in F344 rats. Food Chem

Toxicol. 1987; 25(9): 697-702.

Kang CK, Kim MK, Kim MJ, Jun KI, Kim KW, Park SH, Song KH. Acute respiratory failure

caused by phenytoin overdose. Korean J Intern Med. 2013; 28(6): 736-738.

Kemp JW, Tannhauser M, Swinyard EA. Effect of diphenylhydantoin onhexobarbital sleep

time in mice and rats. Arch Int Pharmacodyn Ther. 1971; 193(1): 37-47.

Keppel Hesselink JM, Kopsky DJ. Phenytoin: 80 years young, from epilepsy to breast cancer,

a remarkable molecule with multiple modes of action. J Neurol (2017a) 264:1617-1621

Keppel Hesselink JM, Kopsky DJ. Phenytoin: neuroprotection or neurotoxicity? Neurol Sci,

2017b, 38: 1137-1141.

Keppel Hesselink JM. Phenytoin repositioned in wound healing: clinical experience spanning

60 years. Drug Discov Today. 2018; 23(2): 402-408.


Kindig D, Garriott ML, Parton JW, Brunny JD, Beyers JE. Diphenylhydantoin is not

genotoxic in a battery of short-term cytogenetic assays. Teratog Carcinog Mutagen. 1992;

12(1): 43-50.

Kokenge R, Kutt H, McDowell F. Neurological sequelae folowing dilantin overdose in a

patient and in experimental animals. Neurology 1965; 15: 823-829.

Komatsu T, Yamazaki H, Asahi S, Gillam EMJ, Guengerich FP, Nakajima M, Yokoi T.

Formation of a dihydroxy metabolite of phenytoin by human liver microsomes/cytosol: Roles

of cytochrome P450 2C9, 2C19, and 3A4. Drug Metab Dispos. 2000; 28: 1361–1368.

Kopsky DJ, Keppel Hesselink JM. Topical phenytoin for the treatment of neuropathic pain. J

Pain Res. 2017; 10: 469-473.

Krishnamoorthy KS, Zalneraitis EL, Young RS, Bernad PG. Phenytoin-induced

choreoathetosis in infancy: case reports and a review. Pediatrics. 1983; 72(6): 831-834.

Kumar N, Chakraborty A, Suresh SH, Basappaji S, Betdur AL. Phenytoin-induced cerebellar

atrophy in an epileptic boy. Indian J Pharmacol. 2013; 45(6): 636-637.

Kumkamthornkul P, Udnaen S, Tansit T, Tuchinda P, Srinoulprasert Y. Evaluation of a

lymphocyte transformation test and cytokine detection assay to identify phenytoin and

carbamazepine provoked DRESS or SJS/TEN in epilepsy patients. Int Immunopharmacol.

2018, 63:204-210

Lakehal F, Wurden CJ, Kalhorn TF, Levy RH. Carbamazepine and oxcarbazepine decrease

phenytoin metabolism through inhibition of CYP2C19. Epilepsy Res. 2002, 52(2):79-83.


Lee SK, Mori S, Kim DJ, Kim SY, Kim SY, Chu M, Heo K, Lee BI, Kim DI. Diffusion

tensor MRI and fiber tractography of cerebellar atrophy in phenytoin users. Epilepsia. 2003;

44(12): 1536-1540.

Lee TJ, Carney CN, Lapis JL, Higgins T, Fallon HJ. Diphenylhydantoin-induced hepatic

necrosis. A case study. Gastroenterology. 1976; 70(3): 422-424.

Léonard A, de Meester C, Fabry L, de Saint-Georges L, Dumont P. Lack of mutagenicity of

diphenylhydantoin in in vitro short-term tests. Mutat Res. 1984; 137(2-3): 79-88.

Lewis RJ. Sr. (ed) Sax's Dangerous Properties of Industrial Materials. 11th Edition. Wiley-

Interscience, Wiley & Sons, Inc. Hoboken, NJ. 2004, p. 1302.

Lide DR, Milne GWA. (eds.). Handbook of Data on Organic Compounds. Volume I. 3rd ed.

CRC Press, Inc. Boca Raton, FL. 1994., p. V4: 3216.

López L, Thomson A, Rabinowicz AL. Assessment of colour vision in epileptic patients

exposed to single-drug therapy. Eur Neurol. 1999; 41(4): 201-205.

Maeda T, Sano N, Togei K, Shibata M, Izumi K, Otsuka H. Lack of carcinogenicity of

phenytoin in (C57BL/6 x C3H)F1 mice. J Toxicol Environ Health. 1988; 24(1): 111-1119.

Magadle R, Weiner P, Waizman J. [Atrioventricular junctional arrhythmia due to oral

phenytoin intoxication]. Harefuah. 1999; 136(6): 446-448, 514. Hebrew.

Maguire JH. Quantitative estimation of catechol/methylcatechol pathways in human

phenytoin metabolism. Epilepsia 1988; 29(6): 753-759.

Mahatma M, Haponik EF, Nelson S, Lopez A, Summer WR. Phenytoin-induced acute

respiratory failure with pulmonary eosinophilia. Am J Med. 1989; 87(1): 93-94.


Martin E, Gambertoglio JG, Adler DS, Tozer TN, Roman LA, Grausz H. Removal of

phenytoin by hemodialysis in uremic patients. JAMA 1977; 238(16): 1750–1753.

Martin LJ, Chao R, Corry B. Molecular dynamics simulation of the partitioning of benzocaine

and phenytoin into a lipid bilayer. Biophys Chem. 2014, 185:98-107.

Masuda Y, Karasawa T, Shiraishi Y, Hori M, Yoshida K, Shimizu M. 3-Sulfamoylmethyl-

1,2-benzisoxazole, a new type of anticonvulsant drug. Pharmacological profile.

Arzneimittelforschung. 1980; 30(3): 477-4783.

Mathews SR, Badyal DK, Mathew R. Phenytoin-induced bradycardia and hypotension. Indian

J Pharmacol. 2019, 51(2):120-122.

Matsue Y, Wagatsuma S, Ito K, Okuma T. Smooth pursuit eye movements in epileptics under

antiepileptic medication. Tohoku J Exp Med. 1981; 134(4): 341-349.

Matsuura M. Epileptic psychoses and anticonvulsant drug treatment. J Neurol Neurosurg

Psychiatry. 1999; 67(2): 231–233.

McLain LW, Martain JT, Allen JH. Cerebellar degeneration due to chronic phenytoin therapy.

Ann Neurol 1980; 7(1): 18-23.

McNamara JO. Drugs effective in the therapy of the epilepsies. In: Hardman JG, Limbird LE,

Molinoff PB, Ruddon RW, editors. Goodman and Gilman’s The Pharmacological Basis of

Therapeutics. 9th Ed. New York: McGrawHill; 1996. pp 461-486.

Mehndiratta S. Phenytoin-induced encephalopathy in a child. Indian J Pharmacol. 2016;

48(4): 460-461.
Merlo González VE, Arbol Linde F, Rodríguez J, López Onega P. [Ventricular fibrillation

during intravenous administration of phenytoin]. An Med Interna. 2002; 19(3): 158-159.

Spanish.

Merritt HH, Putnam TJ. Landmark article Sept 17, 1938: Sodium diphenyl hydantoinate in the

treatment of convulsive disorders. By H. Houston Merritt and Tracy J. Putnam. JAMA. 1984;

251(8): 1062-1067.

Mirkin ML. Diphenylhydantoin: placental transport, fetal localisation, neonatal metabolism,

and possible teratogenic effects. J Pediatr. 1971; 78(2): 329–337.

Montes de Oca-Luna R, Leal-Garza CH, Baca-Sevilla S, Garza-Chapa R. The effect of

diphenylhydantoin on the frequency of micronuclei in bone-marrow polychromatic

erythrocytes of mice. Mutat Res. 1984; 141(3-4): 183-187.

Munns AJ, De Voss JJ, Hooper WD, Dickinson RG, Gillam EMJ. Bioactivation of phenytoin

by human cytochrome P450: Characterization of the mechanism and targets of covalent

adduct formation. Chem Res Toxicol. 1997; 10: 1049–1058.

Nau H, Kuhnz W, Egger HJ, Rating D, Helge H. Anticonvulsants during pregnancy and

lactation. Transplacental, maternal and neonatal pharmacokinetics. Clin Pharmacokinet. 1982;

7(6): 508–543.

Ney GC, Lantos G, Barr WB, Schaul N. Cerebellar atrophy in patients with long term

phenytoin exposure and epilepsy. Arch Neurol 1994; 51(8): 767-771.

Nolan SJ, Marson AG, Pulman J, Tudur Smith C. Phenytoin versus valproate monotherapy

for partial onset seizures and generalised onset tonic-clonic seizures. Cochrane Database Syst

Rev. 2013a; (8): CD001769.


Nolan SJ, Tudur Smith C, Pulman J, Marson AG. Phenobarbitone versus phenytoin

monotherapy for partial onset seizures and generalised onset tonic-clonic seizures. Cochrane

Database Syst Rev. 2013b; (1): CD002217.

Nseir G, Golshayan D, Barbey F. Phenytoin-associated severe hypocalcemia with seizures in

a patient with a TSC2-PKD1 contiguous gene syndrome. Ren Fail. 2013; 35(6): 866-868.

NTP (National Toxicology Program). Toxicology and Carcinogenesis Studies of 5,5-

Diphenylhydantoin (CAS No. 57-41-0) (Phenytoin) in F344/N Rats and B6C3F1 Mice (Feed

Studies). Natl Toxicol Program Tech Rep Ser. 1993; 404: 1-303.

Oelze LL, Pillow MT. Phenytoin-induced drug reaction with eosinophilia and systemic

symptoms (DRESS) syndrome: a case report from the emergency department. J Emerg Med.

2013 ; 44(1): 75-78.

Okogbaa JI, Onor IO, Arije OA, Harris MB, Lillis RA. Phenytoin-Induced Purple Glove

Syndrome: A Case Report and Review of the Literature. Hosp Pharm. 2015; 50(5): 391-395.

Onaolapo AY, Adebayo AA, Onaolapo OJ. Oral phenytoin protects against experimental

cyclophosphamide-chemotherapy induced hair loss. Pathophysiology. 2018; 25(1): 31-39.

O'Neil MJ. (ed.). The Merck Index - An Encyclopedia of Chemicals, Drugs, and Biologicals.

13th Edition, Whitehouse Station, NJ: Merck and Co., Inc., 2001., p. 1312.

Osorio I, Reed RC. Treatment of refractory generalized tonic‐clonic status epilepticus with

pentobarbital anesthesia after high‐dose phenytoin. Epilepsia, 1989; 30(4): 464-471.

Ozkaynakci A, Gulcebi MI, Ergeç D, Ulucan K, Uzan M, Ozkara C, Guney I, Onat FY. The

effect of polymorphic metabolism enzymes on serum phenytoin level. Neurol Sci.

2015,36(3):397-401.
Prasad M, Krishnan PR, Sequeira R, Al-Roomi K. Anticonvulsant therapy for status

epilepticus. Cochrane Database Syst Rev. 2014; (9): CD003723.

Praveen-kumar S, Desai M. Ocular motor abnormalities in a patient with phenytoin toxicity--

case report and minireview. Clin Neurol Neurosurg. 2014; 127: 116-117.

Raftopoulos R, Hickman SJ, Toosy A, Sharrack B, Mallik S, Paling D, Altmann DR,

Yiannakas MC, Malladi P, Sheridan R, Sarrigiannis PG, Hoggard N, Koltzenburg M, Gandini

Wheeler-Kingshott CA, Schmierer K, Giovannoni G, Miller DH, Kapoor R. Phenytoin for

neuroprotection in patients with acute optic neuritis: a randomised, placebo-controlled, phase

2 trial. Lancet Neurol. 2016; 15(3): 259-269.

Razmaraii N, Babaei H, Nayebi AM, Asadnasab G, Helan JA, Azarmi Y. Cardioprotective

effect of phenytoin on doxorubicin-induced cardiac toxicity in a rat model. J Cardiovascular

Pharm. 2016; 67(3): 237-245.

Reynolds EH. Schizophrenia-like psychoses of epilepsy and disturbances of folate and

Vitamin B12 metabolism induced by anticonvulsant drugs. Br J Psychiatry. 1967; 113(501):

911–919.

Riedel L, Obe G. Mutagenicity of antiepileptic drugs. II. Phenytoin, primidone and

phenobarbital. Mutat Res. 1984; 138(1): 71-74.

Sadeghi L, Yekta R, Dehghan G. New mechanisms of phenytoin in calcium homeostasis:

competitive inhibition of CD38 in hippocampal cells. Daru. 2018, 26(2):191-198.

Safari J, Naeimi H, Ghanbari MM, Fini OS. Preparation of phenytoin derivatives under

solvent-free conditions using microwave irradiation. Russian J Org Chem 2009; 45(3): 477-

479.
Salinsky MC, Oken BS, Binder LM. Assessment of drowsiness in epilepsy patients receiving

chronic antiepileptic drug therapy. Epilepsia. 1996; 37(2): 181-187.

Sangster J. LOGKOW Databank. Sangster Res. Lab., Montreal Quebec, Canada, 1994.

Sasaki H, Ikedo D, Kataoka M, Kido JI, Kitamura S, Nagata T. Pronounced Palatal and

Mandibular Tori Observed in Patient With Chronic Phenytoin Therapy: A Case Report. J

Periodontol. 1999; 70(4): 445-448.

Scott JM, Weir DG. Drug-induced megaloblastic change. Clin Haematol. 1980; 9(3): 587-

606.

Scriba GK, Lambert DM, Poupaert JH. Bioavailability of phenytoin following oral

administration of phenytoin-lipid conjugates to rats. J Pharm Pharmacol. 1995, 47(11):945-

948.

Selhorst JB, Kaufman B, Horwitz SJ. Diphenylhyadantoin induced cerebellar degeneration.

Arch Neurol 1972; 27(5): 453-455.

Sezzano P, Raimondi A, Arboix M, Pantarotto C. Mutagenicity of diphenylhydantoin and

some of its metabolites towards salmonella typhimurium strains. Mutat Res. 1982; 103(3-6):

219-228.

Shanmugarajah PD, Hoggard N, Aeschlimann DP, Aeschlimann PC, Dennis GJ, Howell SJ,

Reuber M, Grünewald RA, Hadjivassiliou M. Phenytoin-related ataxia in patients with

epilepsy: clinical and radiological characteristics. Seizure. 2018; 56: 26-30.

Schwinghammer TL, Howrie DL. Phenytoin-induced lymphadenopathy. Drug Intell Clin

Pharm. 1983; 17(6): 460-462.


Silvado CE, Terra VC, Twardowschy CA. CYP2C9 polymorphisms in epilepsy: influence on

phenytoin treatment. Pharmgenomics Pers Med. 2018; 11: 51-58.

Şimşek G, Ciftci O, Karadag N, Karatas E, Kizilay A. Effects of topical phenytoin on nasal

wound healing after mechanical trauma: An experimental study. Laryngoscope. 2014;

124(12) :E449-E454.

Singh G, Driever PH, Sander JW. Cancer risk in people with epilepsy: the role of antiepileptic

drugs. Brain. 2005; 128(Pt 1): 7-17.

Siniscalchi A, Gallelli L, Russo E, De Sarro G. A review on antiepileptic drugs-dependent

fatigue: pathophysiological mechanisms and incidence. Eur J Pharmacol. 2013; 718(1-3): 10-

16.

Smythe MA, Umstead GS. Phenytoin hepatotoxicity: a review of the literature. DICP. 1989;

23(1): 13-18.

Somberg J. Antiarrhythmic drug therapy. Recent advances and current status. Cardiology.

1985; 72(5-6): 329-348.

Srinivasan G, Wyawahare M, Mathen PG, Subrahmanyam DK. Life-threatening

bradyarrhythmia with oral phenytoin overdose. J Pharmacol Pharmacother. 2015; 6(3): 179-

181.

Stephens JH, Shaffer JW. A controlled study of the effects of diphenylhydantoin on anxiety,

irritability, and anger in neurotic outpatients. Psychopharmacologia. 1970; 17(2): 169-181.

Stille G, Brunckow I. [The ganglioplegic effect of 3-alkylaminoalkyl-hydantoins].

Arzneimittelforschung. 1954; 4(12): 723-725. German.


Sudarsan P, Varghese R, Suresh A, Radhakrishnan A. Phenytoin-Induced Paradoxical

Seizures and Blood Dyscrasias in a Patient Receiving Rapid Intravenous Infusion: A Case

Report. J Pharm Pract. 2019, 26:897190019894868. doi: 10.1177/0897190019894868. [Epub

ahead of print]

Sugimoto M, Wakabayashi Y, Shiokawa Y, Takaku F. [Bone marrow heme synthesis

suppression by diphenylhydantoin in vitro in pure red cell aplasia]. Rinsho Ketsueki. 1982;

23(9): 1446-1452. Japanese.

Sullivan JR, Shear NH. The drug hypersensitivity syndrome: what is the pathogenesis? Arch

Dermatol. 2001; 137(3): 357-364.

SunshineI. (ed.). CRC Handbook of Analytical Toxicology. Cleveland: The Chemical Rubber

Co., 1969., p. 44.

Suwalsky M, Mennickent S, Norris B, Cárdenas H. The antiepileptic drug phenytoin affects

sodium transport in toad epithelium. Comp Biochem Physiol C Toxicol Pharmacol. 2006,

142(3-4):253-261.

Szabo GK, Pylilo RJ, Davoudi H, Browne TR. Simultaneous determination of p-hydroxylated

and dihydrodiol metabolites of phenytoin in urine by high-performance liquid

chromatography. J Chromatogr. 1990; 535(1-2): 279-285.

Taing KD, O'Brien TJ, Williams DA, French CR. Anti-epileptic drug combination efficacy in

an in vitro seizure model - phenytoin and valproate, lamotrigine and valproate. PLoS One.

2017, 12(1):e0169974.

Tan EK, Chan LL, Auchus AP. Phenytoin cerebellopathy without epilepsy. Acta Neurol

Scand. 2001; 104(1): 61-62.


Thangaraju P, Tamilselvan T, Venkatesan S, Eswaran T, Singh H, Giri VC, Showkath Ali,

MK. Topical Phenytoin for Managing Various Ulcers:A meta-analysis. Sudan Medical

Monitor. 3015; 10 (2): 63–67.

Theil GB, Richter RW, Powell MR, Doolan PD. Acute dilantin poisoning. Neurology. 1961;

11: 138-142.

Thomson PDR, Micromedex. Drug Information for the Health Care Professional. Volume 1,

Greenwood Village, CO. 2007., p. 271. ISBN-13: 978-1563635144

TOXNET. Data reported in TOXNET (https://chem.nlm.nih.gov/chemidplus/rn/20830-75-5)

has been used. Original literature sources are unthinkable.

Treiman DM, Woodbury DM. Phenytoin: absorption, distribution, and excretion. In; Levy

RH, Mattson RH, Meldrum BS, eds, Antiepileptic Drugs, 4th Ed., New York, Raven Press,

1995; pp. 301–314.

Urashima Y, Urashima K, Ohnishi M, Matsushita K, Suzuki K, Kurachi K, Nishihara M,

Katsumata T, Myotoku M, Ikeda K, Hirotani Y. Interaction between phenytoin and enteral

nutrients and its influence on gastrointestinal absorption. Pharmazie. 2019, 74(9):559-562.

Usdin E, Efron DH, (ed.), Psychotropic Drugs and Related Compounds, 2nd ed., Washington

DC, 1972; Vol. -, p. 224.

Velasco MJ, McDermott J. Drug rash with eosinophilia and systemic symptoms (DRESS)

syndrome and hepatitis induced by phenytoin. Int J Dermatol. 2014; 53(4): 490-493.

Weichbrodt GD, Elliott DP. Treatment of phenytoin toxicity with repeated doses of activated

charcoal. Ann Emerg Med. 1987; 16(12): 1387-1389.


Wilson JT, Höjer B, Rane A. Loading and conventional dose therapy with phenytoin in

children: kinetic profile of parent drug and main metabolite in plasma. Clin Pharmacol Ther.

1976; 20(1): 48-58.

Wu X, Liu W, Zhou W. Association of CYP2C9*3 with phenytoin-induced Stevens-Johnson

syndrome and toxic epidermal necrolysis: A systematic review and meta-analysis. J Clin

Pharm Ther. 2018; 43(3): 408-413.

Yabuki S, Nakaya K. Immunoglobulin abnormalities in epileptic patients treated with

diphenylhydantoin. Folia Psychiatr Neurol Jpn. 1976; 30(2): 93-109.

Yalkowsky SH, He Y. Handbook of Aqueous Solubility Data: An Extensive Compilation of

Aqueous Solubility Data for Organic Compounds Extracted from the AQUASOL

dATAbASE. CRC Press LLC, Boca Raton, FL. 2003., p. 998.

Yamazaki H, Hiroki S, Urano T, Inoue K, Shimada T. Effects of roxithromycin, erythromycin

and troleandomycin on their N-demethylation by rat and human cytochrome P450 enzymes.

Xenobiotica 1996; 26: 1143–1153.

Yamazaki H, Komatsu T, Takemoto K, Saeki M, Minami Y, Kawaguchi Y, Shimada N,

Nakajima M, Yokoi T. Decreases in phenytoin hydroxylation activities ctalyzed by liver

microsomal cytochrome P450 enzymes in phenytoin-trated rats. Drug Metab Disp. 2001;

29(4): 427-434.

Yampayon K, Sukasem C, Limwongse C, Chinvarun Y, Tempark T, Rerkpattanapipat T,

Kijsanayotin P. Influence of genetic and non-genetic factors on phenytoin-induced severe

cutaneous adverse drug reactions. Eur J Clin Pharmacol. 2017; 73(7): 855-865.
Yasumori T, Chen LS, Li QH, Ueda M, Tsuzuki T, Goldstein JA, Kato R, Yamazoe Y.

Human CYP2C-mediated stereoselective phenytoin hydroxylation in Japanese: difference in

chiral preference of CYP2C9 and CYP2C19. Biochem Pharmacol. 1999; 57(11): 1297-1303.

Zhang L, Li Z, Ma G, Han X, Li C, Shan M, Chen LA. Systematic review of phenytoin

intoxication induced by compound phenytoin sodium, ephedrine hydrochloride and

theophylline tablets in China. Medicine (Baltimore). 2018, 97(51):e13689.

Zinsmeister S, Marks RE. Acute athetosis as a result of phenytoin toxicity in a child. Am J

Dis Child. 1976; 130(1): 75-76.

Zuin DR, Neme R, Porta L, Vera J, Lopez OL. [Acute cerebellar atrophy caused by

diphenylhydantoin intoxication resulting from drug interaction]. Rev Neurol. 2003; 36(2):

195-196. Spanish.
Table 1. Published Toxicity Data for Phenytoin

Species Test Type * 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 800mg/kg TOXNET
(800mg/kg)
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

* LD50 = Median Lethal Dose, LDLo = Lethal Dose Low, TDLo = Toxic Dose Low, NOAEL

= No-Observed-Adverse-Effect Level
Figure 1. Chemical structure of phenytoin
Figure 2. Proposed pathways of phenytoin metabolism.
HIGHLIGHTS

• phenytoin chemistry, pharmacokinetics and toxicology is reviewed

• special accent on mutagenicity, carcinogenicity and teratogenicity of phenytoin is

made

• side effects of phenytoin on human health are thoroughly described

• special attention on DRESS syndrome and cerebellar atrophy is paid


Declaration of interests

☒ The authors declare that they have no known competing financial interests or personal relationships
that could have appeared to influence the work reported in this paper.

☒The authors declare the following financial interests/personal relationships which may be considered
as potential competing interests:

Qinghua Wu
Declaration of interests

☒ The authors declare that they have no known competing financial interests or personal relationships
that could have appeared to influence the work reported in this paper.

☒The authors declare the following financial interests/personal relationships which may be considered
as potential competing interests:

Eugenie Nepovimova
Declaration of interests

☒ The authors declare that they have no known competing financial interests or personal relationships
that could have appeared to influence the work reported in this paper.

☒The authors declare the following financial interests/personal relationships which may be considered
as potential competing interests:

Jiri Patocka
Declaration of interests

☒ The authors declare that they have no known competing financial interests or personal relationships
that could have appeared to influence the work reported in this paper.

☒The authors declare the following financial interests/personal relationships which may be considered
as potential competing interests:

Kamil Kuca

You might also like