You are on page 1of 10

960 Send Orders for Reprints to reprints@benthamscience.

ae
Current Drug Metabolism, 2018, 19, 960-969

REVIEWARTICLE
ISSN: 1389-2002
eISSN: 1875-5453

Current
Drug
Hypoxia Plays a Key Role in the Pharmacokinetic Changes of Drugs Metabolism
Impact
Factor:
2.655

at High Altitude The international

journal for timely

in-depth reviews

on Drug Metabolism

BENTHAM
SCIENCE

Xuejiao Zhou1, Yongqiong Nian2, Yijie Qiao3, Meng Yang2, Yuanyao Xin2 and Xiangyang Li1,*
1
State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China; 2Department of Resources of Biology,
College of Eco-Environmental Engineering, Qinghai University, Xining, China; 3Department of Pharmacy, Medical College, Qing-
hai University, Xining, China
Abstract: Background: Hypoxia can alter the Pharmacokinetic (PK) characteristics of drugs, thereby affecting drug
absorption, distribution, metabolism, and excretion. Environmental characteristics at high altitude include but are not
limited to hypobaric hypoxia, low temperature, high solar radiation, and arid climate, all of which can adversely
affect normal bodily functions. Therefore, it is important to study the pharmacokinetic changes of drugs at high altitude.
Method: A systematic review of published studies was carried out to investigate the effects of hypoxia on the meta-
bolic characteristics of some drugs and the activity and expression of drug-metabolizing enzymes in high-altitude
hypoxic environments, and discussed the relevant mechanisms.
ARTICLE HISTORY Results: The metabolism of most drugs decreases in high-altitude hypoxia, whereas Mean Residence Time (MRT),
Received: January 02, 2018 Half Time (T1/2), and Area Under the Curve (AUC) increase and Clearance (CL) decrease in this environment. The
Revised: March 08, 2018 effect of hypoxia on CYP450 enzymes in animals is still a subject of debate. With the exception of CYP2C11 and
Accepted: March 19, 2018 CYP2C22, the widespread belief is that high-altitude hypoxia decreased the activity and expression of CYP1A1,
DOI: CYP1A2, CYP2E1, and CYP3A1, and increased those of CYP3A6 and CYP2D1 in rats. The changes in the activity
10.2174/1389200219666180529112913 and expression of drug metabolizing enzymes are consistent with the changes in pharmacokinetics of some enzyme
substrates in the high-altitude hypoxia environment.
Conclusion: The findings of this review have indicated that hypoxia may play a key role in the PK changes of drugs
Current Drug Metabolism

at high altitude. It is suggested that patient living at or traveling to high altitude should be closely monitored, and the
dosages of some drugs metabolized should be reduced.
Keywords: High altitude, hypoxia, pharmacokinetics, drug metabolism, drug-metabolizing enzyme, mechanism.
1. INTRODUCTION attention has also been paid to the effects of hypoxia on drug me-
Plateaus are large areas of land that usually exceed 1,000 m, tabolism, which are mostly physiological but can also be pathologi-
and are characterized by steep slopes, vast area, and wide terrain. It cal. Both of these effects influence drug absorption, distribution,
is generally recognized that particular physiological responses oc- metabolism, and excretion, leading to changes in the pharmacoki-
cur at above 1,500 m in altitude. Plateau conditions are character- netic (PK) characteristics of drugs [8-10]. Human activities at high
ized by hypoxia, low pressure, low temperature, strong wind, and altitude have increased in recent years; thus, rational drug use in
dry air, all of which can adversely affect normal bodily functions. hypoxic conditions has been the subject of debate.
High-altitude hypoxia is classified into light hypoxia (1,500-2,500 This review summarized the effects of high-altitude hypoxia on
m), moderate hypoxia (2,500-4,500 m), and severe hypoxia (4,500- drug metabolism, elaborated on the metabolic characteristics of
5,500 m) according to sea level height. The higher the altitude, the some drugs and the activity and expression of drug-metabolizing
lower the atmospheric pressure and available oxygen. The decrease enzymes in this environment, and investigated the relevant mecha-
in partial pressure of oxygen in the atmosphere at high altitude, nisms, with the goal of providing information about rational drug
resulted in decreased oxygen dispersed into the blood via pulmo- use at high altitude.
nary alveoli, as well as a decrease in arterial partial pressure and 2. DRUG PHARMACOKINETICS UNDER HYPOXIA
saturation of oxygen, leading to hypoxemia and histanoxia [1].
Hypoxia-induced fluctuations in organism function, metabolism, People traveling to high altitudes for sport, as tourists or for
and structure are the pathological basis of disease at high altitude. military and other reasons are usually healthy and fit, but some-
times use drugs for common ailments like influenza, acute, and
High-altitude hypoxic environments significantly affect the chronic high-altitude sickness. Lower oxygen levels at high altitude
circulatory, nervous, and endocrine systems, as well as metabolism, may compromise the metabolism of drugs. High-altitude hypoxia
which in turn cause changes in bodily functions such as metabolism affects drug PK, which may influence treatment decisions as drug
and growth. This leads to altitude sickness including acute altitude metabolism in vivo requires oxygen. Thus, when the hepatic meta-
stress, pulmonary edema, encephalopathy, chronic polycythemia, bolic capacity of the liver is limited in hypoxic conditions, drug
and cardiovascular disease [2, 3]. Hypoxia causes a range of effects biotransformation and clearance are modified. Powell et al. [11]
on bodily functions and metabolism, and the degree and conse- found that patients with the chronic obstructive pulmonary disease,
quence of which are determined by its speed of entry into the pla- pulmonary edema, pulmonary cardiopathy, or cardiac failure main-
teau, residence time at high altitude, and body metabolic status. A tained a low level of systemic clearance (CL), with a reduction of
slow adjustment to high altitude is typically followed by compensa- 30–60%. These diseases are all hypoxia-related; hence, we specu-
tory reactions, whereas a rapid adjustment gives rise to metabolic lated that decreases in theophylline CL are due to low oxygen lev-
impairment and systemic disease [4, 5]. els. These findings were subsequently confirmed, and the CL of
Studies of high-altitude hypoxia have focused on prevention, theophylline was shown to decrease by 25.7% in rabbits under
nutrition, and adaption [6, 7]. However, since the 1980s, close acute hypoxia [12, 13]. Du Souich [14] found that acute hypoxia did
not alter diltiazem disposition; however, the CL and Volume of
*Address correspondence to this author at the State Key Laboratory of Pla- Distribution (Vd) were significantly decreased in dogs subjected to
teau Ecology and agriculture, Qinghai University, Xining 810016, China. a fraction of inspired oxygen (FiO2) of 8% for 120 h compared with
Tel: (86-971)536 2083. Fax: (86-971)536 2383. E-mail: qhmclxy@163.com dogs breathing 21% FiO2.

 1875-5453/18 $58.00+.00 © 2018 Bentham Science Publishers


Hypoxia Plays a Key Role in the Pharmacokinetic Changes of Drugs at High Altitude Current Drug Metabolism, 2018, Vol. 19, No. 11 961

Acute hypoxia increased the area under the curve (AUC), mean acute and chronic hypoxia, the Cmax of furosemide decreased and the
residence time (MRT), half-life (T1/2), and peak concentration T1/2 and CL/F increased. The MRT, AUC, and Vd for furosemide
(Cmax) of propranolol in rats after exposure to an altitude of 4,010 under acute hypoxia were reduced, but the MRT, AUC, and Vd for
m, but apparent clearance (CL/F) and Vd were significantly de- chronic hypoxia were increased. Acute and chronic high-altitude
creased [15]. High-altitude hypoxia also altered metoprolol disposi- hypoxia increased the AUC of prednisolone by 12.8% and 13.5% and
tion, and the AUC, Vd, and C max were significantly decreased, but the Cmax by 16.9% and 14.1%, respectively. On the other hand, acute
the MRT, CL/F, and T1/2 were significantly increased in rats after and chronic high-altitude hypoxia decreased the Vd by 20.4% and
exposure to an altitude of 4,010 m [16]. When rats ascended to 15.6% and the CL/F by 25.2% and 15.6%, respectively. A study on
4,300 m, the AUC, MRT, and Cmax of amoxicillin significantly lithium carbonate demonstrated that the T1/2 increased by 64.1% and
increased, but the CL/F was significantly reduced [17]. Anjana et 111.4% in volunteers after acute and chronic exposure to high alti-
al. [18] found that the T1/2 of gentamicin, acetlysalicylic acid, aceta- tude, respectively, compared to volunteers residing at low altitude. It
zolamide, and phenobarbitone was increased in rabbits through was also found that the Vd increased by 18.9% and 35.8% and the
simulations of acute hypoxia, which was equivalent to 4,572 m in MRT increased by 79.3% and 139.6% in acute and chronic hypoxia
altitude, whereas the elimination rate constant (Ke) decreased for groups, respectively. On the other hand, acute and chronic high- alti-
each drug. The MRT and T1/2 of ibuprofen in rats were significantly tude hypoxia decreased CL/F by 24.3% and 37.3%, respectively [23].
increased by 42% and 51%, respectively, at an altitude equivalent A study that evaluated the effects of high altitude on meperidine
to 7,620 m above sea level [19]. Du Souich [20] studied the effects demonstrated that the CL/F was significantly lower and the T1/2 and
of hypercapnia and/or hypoxemia on the kinetics and concentra- MRT were significantly higher at high altitudes than at sea level [24].
tions of phenytoin in the cerebrospinal fluid of conscious rabbits One clinical study suggested that, in addition to age, sex, diet, fitness
and found that the CL of phenytoin decreased by 36.9% and the level, the length and degree of hypoxia also contribute to high-
AUC increased by 67.6% in hypoxemic rabbits. altitude hypoxia-induced drug metabolic changes [10].
Due to differences in drug-metabolizing enzymes, metabolic Racial differences can influence the safety, efficacy, dose, and
pathways, and drug metabolites between animals and humans, incon- dosage regimen of drugs, and in high-altitude hypoxic environ-
sistent results can be obtained when studying metabolism in a high- ments, additional differences can influence these drug characteris-
altitude hypoxic environment. Some drugs decrease CL and others tics. Our study showed that the AUC of sulfamethoxazole was sig-
increase CL; in fact, even the same drug can behave differently in nificantly lower in native Tibetan than in native Han subjects living
conditions of acute and chronic hypoxia. After volunteers from the at high altitude, but other PK parameters were not significantly
plain ascended to 4,500 m, the CL of theophylline and verapamil was different between the two groups [25, 26]. The PK parameters of
unchanged, suggesting that acute hypoxia had no effect on either drug lidocaine hydrochloride were significantly changed in native Han
[21]. However, our group found significant changes in the disposition and Tibetan subjects living at high altitude compared to Han sub-
of sulfamethoxazole in healthy male subjects after acute or chronic jects living at low altitude. The T1/2, Vd, and AUC were higher in
exposure to an altitude of 3,780 m compared to those residing at an the two high-altitude groups than in the low altitude group. How-
altitude of 400 m. The T1/2 was 11.5% and 19.9% higher in the acute ever, there was no significant difference between native Han and
and chronic exposure groups, respectively, compared with the low Tibetan subjects living at high altitude [27].
altitude group. The CL/F was 17.8% and 8.9% lower in the acute and In brief, the metabolism of most drugs decreases in high-
chronic exposure groups, respectively, compared with the low alti- altitude hypoxia, whereas the MRT, T1/2, and AUC increase and the
tude group [8]. Arancibia et al [9, 22] studied the PK of furosemide Ke and CL decrease in this environment. Table 1 shows the
and prednisolone in volunteers from the plain, who had ascended to changes in PK of different drugs at hypoxia.
3600 m and lived there for 6 months. The data indicated that for both

Table 1. Changes in pharmacokinetics of different drugs at hypoxia.

Drug; Dosing
Species Model; Method Used Effects References
Route

Theophylline; acute exposure to air (con-


CL decreased from 1.52 ± 0.05 ml/min/kg in control to 1.13 ± 0.13 in hy-
trol) or to a high CO2 and/or
Rabbit Intravenous percapnia, 1.09 ± 0.09 in hypoxemia, and 1.02 ± 0.02 in hypoxemia com- [13]
low O2 atmosphere for 570
injection bined with hypercapnia.
min; HPLC.

Diltiazem; acute or chronic exposure to Acute hypoxia did not alter diltiazem disposition; chronic hypoxia reduced
Dog Intravenous a FiO2 of 8% for 1 h or 120 diltiazem CL from 64 ± 3 to 51 ± 5 ml/ min/kg, and Vd decreased from 11.4 [14]
injection h; HPLC. ± 1.2 to 9.1 ± 0.3 L.

AUC increased from 11.17 ± 1.38 μg·h/L to 60.61 ± 6.31 after acute expo-
Propranolol; acute exposure to high
sure to high altitude; T1/2 increased from 0.67 ± 0.11 h to 1.09 ± 1.53; MRT
altitude (4,010 m) from low
Rat Oral administra- increased from 0.92 ± 0.10 h to 1.44 ± 0.24; CL/F decreased from 141.49 ± [15]
altitude (50 m); LC-
tion 17.30 L/h/kg to 27.06 ± 3.69; Vd decreased from 131.44 ± 11.05 L to 40.82
MS/MS.
± 4.00; Cmax increased from 10.61 ± 0.93 μg/L to 48.06 ± 11.91.

AUC decreased from 6.94 ± 0.79 μg·h/ml to 5.87 ± 1.01 after acute expo-
Metoprolol; acute exposure to high
sure to high altitude; T1/2 increased from 1.25 ± 0.33 h to 2.97 ± 1.53; MRT
altitude (4,010 m) from low
Rat Oral administra- increased from 2.14 ± 0.11 h to 3.16 ± 0.32; CL/F increased from 0.56 ± [16]
altitude (50 m); LC-
tion 0.06 L/h/kg to 1.75 ± 0.28; Vd decreased from 1.00 ± 0.29 L to 0.67 ± 0.11;
MS/MS.
Cmax decreased from 2.49 ± 0.26 μg/ml to 1.60 ± 0.31.

Table (1) contd….


962 Current Drug Metabolism, 2018, Vol. 19, No. 11 Zhou et al.

Drug; Dosing
Species Model; Method Used Effects References
Route

Amoxicillin; acute exposure to high AUC increased from 11.62 ± 1.07 μg·h/ml to 47.878 ± 9.42 after acute expo-
altitude (4,300 m) from low sure to high altitude; T1/2 increased from 1.12 ± 0.12 h to 1.31 ± 0.23; MRT
Rat Oral administra- [17]
altitude (50 m); LC- increased from 1.51 ± 0.12 h to 2.54 ± 0.44; CL/F decreased from 1.02 ± 0.09
tion MS/MS. L/h/kg to 0.26 ± 0.05; Vd decreased from 1.64 ± 0.20 L to 0.47 ± 0.08.

exposure to hypobaric hy-


Gentamicin; poxia at 42.9 mm Hg pres- T1/2 increased from 1.80 ± 0.12 h to 1.11 ± 0.65; Ke decreased from 0.909 ±
Rabbit intraperitoneal sure equivalent to 4,572 m 0.051 to 0.389 ± 0.041; Vd increased from 163.9 ± 13.9 ml/ kg to 241.4 ± [18]
injection altitude for 5 h/day for 7 12.4; CL decreased from 93.3 ±10.9ml/kg/h to 83.9 ± 6.2.
days; HPLC.

Acetlysalicylic exposure to hypobaric hy-


poxia at 429 mm Hg pres- T1/2 increased from 5.69 ± 1.02 h to 7.63 ± 0.84; Ke decreased from 0.144 ±
acid;
Rabbit sure equivalent to 4,572 m 0.032 to 0.095 ± 0.01; Vd decreased from 1071.1 ± 125.1 ml/ kg to 806.1 ± [18]
intraperitoneal 122.1; CL decreased from 155.2 ± 39.4 ml/kg/h to 72.3 ± 7.0.
altitude for 5 h/day for 7
injection
days; HPLC.

exposure to hypobaric hy-


Acetazolamide; poxia at 429 mm Hg pres-
T1/2 increased by 28%; Ke decreased from 0.720 ± 0.188 to 0.527 ± 0.028;
Rabbit intraperitoneal sure equivalent to 4,572 m [18]
Vd increased; CL increased.
injection altitude for 5 h/day for 7
days; HPLC.

exposure to hypobaric hy-


Pentobarbital; poxia at 429 mm Hg pres- T1/2 increased from 7.28 ± 3.33 h to 8.01 ± 2.40; Vd decreased from 950.1 ±
Rabbit intraperitoneal sure equivalent to 4,572 m 178.5 ml/kg to 760.8 ± 238.4; CL decreased from 96.7 ± 27.2 ml/kg/h to [18]
injection altitude for 5 h/day for 7 66.6 ± 10.9.
days; HPLC.

exposure to simulated
Ibuprofen; altitude of 7,620 m for 6 and
Rat T1/2 and MRT increase by 42% and 51% respectively. [19]
Oral administration 24 h in a decompression
chamber; HPLC.

Phenytoin;
exposure to hypoxia at 48 CL decreased from 4.20 ± 0.55 ml/kg/min to 2.65 ± 0.44; AUC increase
Rabbit Intravenous injec- [20]
mm Hg pressure; HPLC. from 2575 ± 319 μg·min/ml to 4316 ± 740.
tion

acute exposure to hypoxia


Theophylline; (12% oxygen correspond-
Human Intravenous injec- ing to an altitude of 4,500 m Acute hypoxia did not alter the pharmacokinetics of theophylline. [21]
tion above sea level); LC-
MS/MS.

acute exposure to hypoxia


Verapamil; (12% oxygen correspond-
Human Intravenous injec- ing to an altitude of 4,500 m Acute hypoxia did not alter the pharmacokinetics of verapamil. [21]
tion above sea level); LC-
MS/MS.

acute high-altitude hypoxia: T1/2 increased from 9.30 ± 1.11 h to 10.37 ±


0.88; MRT increased from 12.06 ± 0.94 h to 13.15 ± 0.67; CL/F decreased
Sulfamethoxa- acute and chronic exposure from 1.01 ± 0.22 L/kg/h to 0.83 ± 0.13; AUC increased from 1202.5 ±
zole; to high altitude (3,800 m) 238.3 μg·h/ml to 1416.3 ± 202.6.
Human [8]
Oral administra- from low altitude (400 m) chronic high-altitude hypoxia: T1/2 increased from 9.30 ± 1.11 h to 11.15 ±
tion for 16 h and 1 year; HPLC. 1.53; MRT increased from 12.06 ± 0.94 h to 13.00 ± 1.01; CL/F decreased
from 1.01 ± 0.22 L/kg/h to 0.92 ± 0.22; AUC increased from 1202.5 ±
238.3 μg·h/ml to 1298.5 ± 256.0.

Table (1) contd….


Hypoxia Plays a Key Role in the Pharmacokinetic Changes of Drugs at High Altitude Current Drug Metabolism, 2018, Vol. 19, No. 11 963

Drug; Dosing
Species Model; Method Used Effects References
Route

acute high-altitude hypoxia: Cmax decreased from 3.93 ± 1.39 μmol/l to 3.54
± 1.35; T1/2 increased from 0.90 ± 0.20 h to 1.13 ± 0.47; CL/F increased
from 0.18 ± 0.04 L/kg/h to 0.20 ± 0.05; MRT decreased from 10.65 ± 4.44
acute and chronic exposure h to 9.62 ± 4.08; AUC decreased from 10.18 ± 2.39 μmol·h/l to 9.23 ± 1.91;
Furosemide;
to high altitude (3,600 m) Vd decreased from 5.05 ± 1.89 L/kg to 4.60 ± 1.01.
Human Oral administra- [22]
from sea level for 15 h and chronic high-altitude hypoxia: Cmax decreased from 3.93 ± 1.39 μmol/l to
tion
6 months; HPLC. 3.70 ± 1.82; T1/2 increased from 0.90 ± 0.20 h to 1.32 ± 0.52; CL/F in-
creased from 0.18 ± 0.04 L/kg/h to 0.19 ± 0.11; MRT increased from 10.65
± 4.44 h to 13.89 ± 10.37; AUC increased from 10.18 ± 2.39 μmol·h/l to
11.22 ± 4.52; Vd increased from 5.05 ± 1.89 L/kg to 5.18 ± 2.27.

acute high-altitude hypoxia: AUC increased from 3.12 ± 0.55 μg·h/ml to


3.52 ± 0.32; Cmax increased from 0.71 ± 0.11 μg/ml to 0.83 ± 0.09; CL/F
Prednisolone; acute and chronic exposure decreased from 440.84 ± 82.88 ml/kg/min to 381.21 ±35.08; Vd decreased
Oral administra- to high altitude (3,600 m) from 1.24 ± 0.27 L/kg to 1.03 ± 0.07.
Human [9]
tion from sea level for 15 h and chronic high-altitude hypoxia: AUC increased from 3.12 ± 0.55 μg·h/ml to
6 months; HPLC. 3.54 ± 0.45; Cmax increased from 0.71 ± 0.11 μg/ml to 0.81 ± 0.06; CL/F
decreased from 440.84 ±82.88 ml/kg/min to 381.42 ± 43.94; Vd decreased
from 1.24 ± 0.27 L/kg to 0.99 ± 0.13.

acute high-altitude hypoxia: T1/2 increased from 9.86 ± 2.21 h to 16.24 ±


acute and chronic exposure 5.64; MRT increased from 10.84 ± 2.68 h to 19.44 ± 7.30; Vd increased
Lithium carbon- to high altitude (4,360 m) from 0.539 ± 0.033 L/kg to 0.641 ± 0.092; CL/F decreased from 0.659 ±
ate; from low altitude (600 m) 0.154 ml/kg/min to 0.499 ± 0.160.
Human [23]
Oral administra- for 15 h and 10 months; chronic high-altitude hypoxia: T1/2 increased from 9.86 ± 2.21 h to 20.84 ±
tion Atomic absorption 3.05; MRT increased from 10.84 ± 2.68 h to 25.97 ± 5.11; Vd increased
spectrophotometry. from 0.539 ± 0.033 L/kg to 0.732 ± 0.130; CL/F decreased from 0.659 ±
0.154 ml/kg/min to 0.413 ± 0.088.

acute high-altitude hypoxia: T1/2 increased from 3.95 ± 0.62 h to 5.09 ±


acute and chronic exposure 1.08; MRT increased from 6.03 ± 0.92 h to 7.75 ± 1.64; CL/F decreased
Meperidine; to high altitude (4,360 m)
from 16.19 ± 3.74 ml/kg/min to 13.05 ± 1.74.
Human intramuscular from sea level for 7 days [24]
and 10 months; Gas chro- chronic high-altitude hypoxia: T1/2 increased from 3.95 ± 0.62 h to 4.80 ±
injection
0.93; MRT increased from 6.03 ± 0.92 h to 7.48 ± 1.29; CL/F decreased
matography.
from 16.19 ± 3.74 ml/kg/min to 12.10 ± 2.28.

native Han volunteers: T1/2 increased from 1.88 ± 0.32 h to 2.44 ± 0.52; Vd
Chinese Han volunteers increased from 3.82 ± 1.01 L/kg to 4.49 ± 0.90; AUC increased from 6.75 ±
living at low altitude (400 1.23 μg·h/ml to 7.08 ± 1.32; CL/F decreased from 1.40 ± 0.29 L/kg/h to
Lidocaine;
m) and in native Han and 1.30 ± 0.27.
Human Oral administra- [27]
Tibetan Chinese volunteers native Tibetan volunteers: T1/2 increased from 1.88 ± 0.32 h to 2.44 ± 0.69;
tion
living at high altitude (2,200 Vd increased from 3.82 ± 1.01 L/kg to 4.31 ± 1.48; AUC increased from
-4,500 m); HPLC. 6.75 ± 1.23 μg·h/ml to 7.43 ± 1.86; CL/F decreased from 1.40 ± 0.29 L/kg/h
to 1.24 ± 0.34.

drug metabolism, is mainly metabolized by CYP1A. Studies on the


3. THE ACTIVITY AND EXPRESSION OF DRUG-
effects of hypoxia on drug-metabolizing enzymes begin with an
METABOLIZING ENZYMES UNDER HYPOXIA
understanding of the CYP450 enzyme system. Proulx & Du Souich
3.1. Cytochrome P450 [30] found that the CYP450 concentration decreased while the ac-
Cytochrome P450 (CYP450) enzymes are components of an tivity remained unchanged in rabbits after acute hypoxia for 8 h, but
oxidase system encoded by a large gene family. Its main subtypes both the concentration and activity decreased after acute hypoxia
include CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, for 24 h, indicating that the effects of hypoxia on CYP450 enzyme
CYP2C19, CYP2D6, CYP2E1, CYP3A4, and CYP3A5. More than activity were time-dependent. Kurdi et al [31] simulated high-
90% of drugs are metabolized by CYP1A2, CYP2C9, CYP2C19, altitude hypoxia with a hypobaric chamber, and found that the ac-
CYP2D6, CYP2E1, and CYP3A4. Among them, CYP3A4 is the tivity and expression of CYP450 from rabbits clearly changed after
body’s most important drug-metabolizing enzyme, biotransforming acute hypoxia for 48 h. CYP1A1 activity significantly decreased,
more than 50% of common drugs [28, 29]. Theophylline, one of the the protein expression of CYP1A1 and CYP1A2 decreased by 20%,
earliest studied drugs for understanding hypoxia-induced changes in and that of CYP3A6 increased by 70% [31]. Under the same ex-
964 Current Drug Metabolism, 2018, Vol. 19, No. 11 Zhou et al.

perimental conditions, Fradette et al. [32, 33] found that acute hy- CYP2E1 and CYP3A1 in rats, which remained unchanged in acute
poxia downregulated CYP1A1, CYP1A2, CYP2B4, CYP2C5, and hypoxia, but significantly decreased in chronic hypoxia. Specifi-
CYP2C16, and upregulated CYP3A6 in rabbit liver. cally, at 2,800 m, chronic hypoxia decreased the activity, protein
We investigated the effects of acute and chronic hypoxia on the expression, and mRNA expression of CYP2E1 by 60.2%, 33.9%,
activity and expression of CYP1A2, CYP2D1, CYP2C11, and 56.0%, respectively, and by 44.2%, 35.5% and 52.0%, respec-
CYP2C22, and N-acetyltransferase 2 (NAT2) in rats, a high- tively at 4,300 m. Chronic hypoxia decreased the activity, protein
altitude environment after either acute or chronic exposure to an expression, and mRNA expression of CYP3A1 by 43.2%, 39.7%
altitude of 3,780 m compared to 400 m. This research group simu- and 31.1% in rats, respectively at 2,800 m, and by 50.1%, 27.5%,
lated a real-life environment and showed that acute or chronic ex- and 31.7%, respectively at 4,300 m [34, 35]. The expression of
posure to high-altitude hypoxia led to a decrease in CYP1A2 activ- CYP2C11 and CYP2C22 was not affected by hypoxia in rats,
ity and expression. Specifically, at 2,800 m, CYP1A2 activity whereas the activity of CYP2C22 slightly increased to 117.6%
dropped by 62.3% and 60.8%, protein expression decreased by when exposed to acute hypoxia of 4,300 m [34, 36]. Studies on
60.4% and 62.0%, and mRNA expression decreased by 51.1% and CYP2D1, CYP2E1, and CYP3A1 have shown that chronic hypoxia
32.9% during acute and chronic exposure, respectively. At 4,300 m, at high altitude plays a key role in enzyme activity and expression.
CYP1A2 activity decreased by 60.8% and 53.8% after acute or The aforementioned results demonstrate that the impact of hypoxia
chronic exposure to high altitude, protein expression decreased by on CYP450 enzymes in animals is still a subject of debate. With the
65.8% and 64.8%, and mRNA expression decreased by 37.2% and exception of CYP2C11 and CYP2C22, the widespread belief is that
30.7% [34]. A study on CYP2D1 showed that at altitudes of 2,800 high-altitude hypoxia decreased the activity and expression of
m and 4,300 m, both the activity and expression of CYP2D1 in- CYP1A1, CYP1A2, CYP2E1, and CYP3A1, and increased those of
creased in rats in chronic hypoxia, whereas CYP2D1 remained CYP3A6 and CYP2D1 in rats (Table 2).
unchanged in acute hypoxia. Similar observations were made for

Table 2. Changes in the activity and expression of drug metabolizing enzymes at hypoxia.

Drug Metabo-
Species Model; Method Used Effects References
lism Enzymes

acute exposure to a FiO2 of 12%


After 8 h of hypoxia, total amount but not the activity of cytochrome
for 8 h or 12 h. Dithionite-
Rabbit CYP450 P450 was decreased; after 24 h of hypoxia, both the amount and the [30]
reduced carbon monoxide dif-
activity of cytochrome P450 were decreased.
ference spectrum.

Rabbit CYP1A1 CYP1A1 protein expression decreased by about 20%.


acute exposure to a FiO2 of 10%
Rabbit CYP1A2 CYP1A2 protein expression decreased by about 20%. [31]
for 24 h. Western blot.
Rabbit CYP3A6 CYP3A6 protein expression increased by about 70%.

Rabbit CYP1A1 CYP1A1 protein expression decreased by 37%.

Rabbit CYP1A2 CYP1A2 protein expression decreased by 40%.

Rabbit CYP2B4 acute exposure to a FiO2 of 8% CYP2B4 protein expression decreased by 55%. [32]
Rabbit CYP2C5 for 48 h. Western blot. CYP2C5 protein expression decreased by 75%.

Rabbit CYP2C16 CYP2C16 protein expression decreased by 82%.

Rabbit CYP3A6 CYP3A6 protein expression increased by 71%.

acute exposure to middle alti- CYP1A2 activity decreased by 62.3%, 60.8%, 60.8% and 53.8% in
tude of 2,800 m from low alti- the AMH, CMH, AHH and CHH groups, respectively; CYP1A2
tude of 400 m for 1 day (AMH); protein expression decreased by 60.4%, 62.0%, 65.8% and 64.8% in
Rat CYP1A2
chronic exposure to middle the AMH, CMH, AHH and CHH groups, respectively; CYP1A2
altitude of 2,800 m from low mRNA expression was decreased 51.1%, 32.9%, 37.2% and 30.7% in
altitude of 400 m for 30 days the AMH, CMH, AHH and CHH groups, respectively.
(CMH); acute exposure to high CYP2D1 activity increased by 86.2% and 176.1% in the CMH and [34]
altitude of 4,300 m from low CHH groups, respectively; CYP2D1 protein expression increased by
Rat CYP2D1 altitude of 400 m for 1 day 20.3% and 23.5% in the CMH and CHH groups, respectively;
(AHH); chronic exposure to CYP2D1 mRNA expression increased by 34.7% in the CHH groups.
high altitude of 4,300 m from
low altitude of 400 m for 30 hypoxia did not affect the activity, protein and mRNA expression of
Rat CYP2C11
days (CHH). HPLC; ELISA; CYP2C11.
real-time PCR.
Rat CYP2C22 CYP2C22 activity increased by 117.6% in the AHH group.

Table (2) contd….


Hypoxia Plays a Key Role in the Pharmacokinetic Changes of Drugs at High Altitude Current Drug Metabolism, 2018, Vol. 19, No. 11 965

Drug Metabo-
Species Model; Method Used Effects References
lism Enzymes

acute exposure to middle alti- CYP2E1 activity decreased by 60.2% and 44.2% in CMH and CHH,
tude of 2,800 m from low alti- respectively; CYP2E1 protein expression decreased by 33.9% and
Rat CYP2E1
tude of 400 m for 1 day (AMH); 35.5% in CMH and CHH, respectively. CYP2E1 mRNA expression
chronic exposure to middle decreased by 56.0% and 52.0% in CMH and CHH, respectively.
altitude of 2,800 m from low [35]
CYP3A1 activity decreased by 43.2% and 50.1% in CMH and CHH,
altitude of 400 m for 30 days
respectively; CYP3A1 protein expression decreased by 39.7% and
Rat CYP3A1 (CMH); acute exposure to high
27.5% in CMH and CHH, respectively; CYP3A1 mRNA expression
altitude of 4,300 m from low
decreased by 31.1% and 31.7% in CMH and CHH, respectively.
altitude of 400 m for 1 day
(AHH); chronic exposure to
high altitude of 4,300 m from NAT2 activity decreased by 23.1% and 28.6% in AMH and AHH,
Rat NAT2 low altitude of 400 m for 30 respectively; NAT2 expression decreased by 55.8% and 19.5% in [34]
days (CHH). HPLC; ELISA; AMH and AHH, respectively.
real-time PCR.

exposure to high altitude of


Rat NAT2 4,600 m from low altitude of NAT2 activity decreased by 38.7%. [10, 38]
400 m for 7 days. HPLC.

exposure to acute hypobaric GST activity decreased by 15% and 23% after 6 h and 24 h of acute
Rat GST [19]
hypoxia for 6 h and 24 h at a hypoxia, respectively.
simulated altitude of 7,620 m.
Rat UGT Fluorimeter. Acute hypoxia did not affect the activity of UGT. [19, 39]

Human CYP1A2 Hypoxia did not affect the activity of CYP1A2.

Human CYP2C19 exposure to acute hypoxia for 7 Hypoxia did not affect the activity of CYP2C19.
days at 4559 m above sea level. [37]
Human CYP2D6 HPLC. a small decrease in the activity of CYP2D6.

Human CYP3A4 a small decrease in the activity of CYP3A4.

There are differences in certain enzymes between humans and drugs should be reduced in the high-altitude hypoxia environment
animals. Jurgens et al. [37] found that the activities of CYP2D6 and [40].
CYP3A4 slowed down after plain volunteers went to 4,559 m pla- Table 2 shows the changes in the activity and expression of
teau and quickened after they returned to a flat area. The activities drug-metabolizing enzymes at hypoxia.
of CYP1A2 and CYP2C19 remained stable throughout this experi-
ment. 4. REGULATION OF CYTOKINES AND NUCLEAR RE-
CEPTORS ON DRUG-METABOLIZING ENZYMES IN HY-
3.2. Other Drug-Metabolizing Enzymes POXIA
Studies on the effects of high-altitude hypoxia on CYP450 en- Studies on the mechanisms underlying the influence of hypoxia
zymes and other drug-metabolizing enzymes have only been re- on drug metabolism are undergoing. High altitude hypoxia can lead
ported in recent years. It has been demonstrated that NAT2 activity to a broad range of physiological changes, leading to the changes in
in rats decreased by 38.7% after they quickly ascended to 4,600 m the ADME process of drugs. It was previously believed that hy-
from 400 m [10, 38]. Subsequent studies further demonstrated that poxia caused blood redistribution in organs, resulting in a decrease
acute high-altitude hypoxia significantly reduces the activity and in hepatic blood flow and inability of the liver to properly break
expression of NAT2 in rats, whereas chronic hypoxia does not have down and utilize drugs, thereby slowing down drug CL [41]. Du
these effects. The activity and mRNA expression of NAT2 in rats Souich et al. [42] found that the CL of lidocaine metabolites de-
under acute hypoxia at 2,800 m decreased by 23.1% and 55.8%, creased in dogs under acute and chronic hypoxia without changes in
respectively, and by 28.6% and 19.5%, respectively at 4,300 m hepatic and kidney blood flow, indicating the lack of a causal rela-
[34]. Shefali et al. [19] simulated a 7,620 m plateau with low cabin tionship between them in hypoxia. Red blood cells, hemoglobin,
pressure and found that the activity of glutathione s-transferase hematocrit and so on are changed in the high altitude hypoxia envi-
(GST) in rats after 6 h and 24 h of acute hypoxia dropped by 15% ronment, and changes in blood cells are closely related to emplace-
and 23%, respectively. Under hypoxia, the activity of UDP- ment time and altitude. It has been claimed that red blood cells are
glucuronsyltransferase (UGT) remains unchanged [19, 39]. increased after exposure to high altitude. Li et al. [8] and Arancibia
The changes in the activity and expression of drug metabolizing et al. [9] found that the hematocrit was significantly increased in
enzymes are consistent with the changes in the pharmacokinetics of healthy subjects after chronic exposure to high altitude, and the
some enzyme substrates in the high-altitude hypoxia environment. binding of drug to red blood cells was significantly higher in the
For example, hypoxia reduces the activity and expression of chronic exposure group than in the low altitude group. The in-
CYP1A2, CYP2B6, CYP2C11, CYP2C22, and NAT2, and the creased binding in the chronic exposure group might be explained
metabolism of their substrates theophylline, phenytoin, ibuprofen, by the increased hematocrit levels. The increased binding to red
and sulfamethoxazole is slow, suggesting that the dosage of these blood cells may also explain the lower clearance observed in the
966 Current Drug Metabolism, 2018, Vol. 19, No. 11 Zhou et al.

groups exposed to a high altitude. Currently, many mechanistic hypoxia, which allows its stabilization [54]. Fisher et al. [55] re-
studies are focused on the effects of hypoxia on cytokines and nu- vealed that the level of human ARD1 protein does not decrease in
clear receptors. hypoxia, nor does HIF-1 acetylation. It remains unknown if these
studies can explain the differences between human and animal drug
4.1. Cytokines metabolism in hypoxia. Rahman & Thomas [56] postulated that the
The effects of hypoxia are similar to those of acute inflamma- downregulation of CYP1A expression is related to IL-1 and nitric
tory reactions, except that the inflammatory response causes down- oxide accumulation. Moreover, regulation of hypoxia is related to
regulation of CYP3A whereas hypoxia causes upregulation [43]. In the activation of P-glycoprotein and AP-1 [33].
rabbits with inflammation, researchers found that interleukin 6 (IL-
6) was the blood modifier for the decrease in CYP450 enzyme ac- 4.2. Nuclear Receptor
tivity. IL-1, Tumor Necrosis Factor (TNF), and Interferon  (IFN- There have been significant improvements in our understanding
) function in weak analogous manners [44-46]. Hypoxia promotes of how exogenous compounds affect CYP450-dependent metabo-
the release of cytokines IL-1, TNF, IFN, Erythropoietin (EPO), lism. Breakthrough discovery was achieved about the inducement
and hypoxia-inducible factor 1 (HIF-1) [47-49], and activates the mechanism of exogenous compounds on liver CYP450 isozyme
protein tyrosine kinase (PTK), protein kinase C (PKC), and mito- expression, which confirmed the role of three main receptors in the
gen-activated protein kinase (MAPK) signal transduction pathways. nuclear receptor superfamily: Pregnane X Receptor (PXR), Consti-
Hypoxia activates HIF-1 and activator protein-1 (AP-1), and their tutive Androstane Receptor (CAR), and the peroxisome prolifera-
translocation to the nucleus and regulates CYP450 enzyme activity tor-activated receptor (PPAR). Studies have shown that PXR is a
and expression [33, 50]. One study showed that after 24 h of serum key inducer of CYP3A expression, whereas CAR mainly regulates
incubation, rabbits in acute hypoxia showed a decrease in CYP1A1 the transcription of CYP2B, CYP1A, and CYP3A [57, 58]. Gene
and CYP1A2 activity and expression, and an increase in the activity knockdown studies have demonstrated that PXR and CAR regulate
and expression of CYP3A6. When antibodies against IFN- and IL- CYP2C9, CYP2C19, and CYP2C18 expression [59, 60]. In addi-
1 were added to the incubation system, the downregulation of tion, most phase II UGT1A drug-metabolizing enzymes are regu-
CYP1A1 and CYP1A2 was suppressed, indicating that IL-1, IL- lated by PXR, with the exception of UGT1A1, which is co-
2, and IFN- regulate their downregulation. The up-regulation of regulated by PXR and CAR [61]. Hypoxia can decrease the expres-
CYP3A6 is influenced by EPO [32]. In addition, hypoxia activates sion of PXR and CAR, thereby further regulating CYP450 activity
the binding of HIF-1 to the CYP3A6 oligonucleotide probe and and expression [62, 63]. Fig. (1) shows the regulatory mechanisms
regulates its activity [33]. Hypoxia improves blood circulation and underlying the drug metabolism of serum cytokines and nuclear
cell metabolism through the regulation of various genes such as receptor in hypoxia. Additional studies are needed to determine the
HIF-1 [51, 52]. HIF-1 consists of  and  subunits, the latter of mechanisms underlying the effects of hypoxia on drugs metabo-
which is the aryl hydrocarbon receptor nuclear translocator, which lized by CYP1A1, CYP1A2, and CYP3A6, among others.
is unaffected by hypoxia. This subunit forms a dimer with the Aryl
Hydrocarbon Receptor (AhR), and translocate to the nucleus where CONCLUSION AND FUTURE OUTLOOKS
it combines and activates its corresponding target genes [41]. The Several previous studies have indicated that hypoxia plays a
downregulation of CYP1A1 and CYP1A2 in hypoxia is related to key role in the PK changes of drugs at high altitude. The metabo-
HIF-1 expression. The expression level of HIF-1 is too low to lism of most drugs decreases in high-altitude hypoxia, with the
detect in aerobic conditions but rapidly increases in hypoxia, after MRT, T1/2, and AUC increase and the Ke and CL decrease. These
which it forms a dimer with HIF-1, inhibiting formation of the findings may have important clinical implications. It is suggested
HIF-1/AhR dimer. AhR is constitutively active in the body, and that patient living at or traveling to high altitude should be closely
CYP1A1 and CPY1A2 are constitutively expressed. HIF-1 is acti- monitored, and the dosages of some drugs metabolized by
vated in hypoxia, inhibiting formation of the HIF-1/AhR het- CYP1A1, CYP1A2, CYP2E1 or CYP3A1 should be reduced.
erodimer and CYP1A expression [50, 53]. The ARD1 of mammals Travel to high altitude can lead to medical problems, from the
can help HIF-1 acetylation and degradation, but is inhibited in mild symptoms of acute mountain sickness to the potentially fatal

O2 IL-1β IL-2β IFN-γ

PTK
PXR PKC MAPKs
ROI
CAR

HIF-1 AP-1

DNA

mRNA

CYP450

Fig. (1). The regulatory mechanisms underlying the drug metabolism of serum cytokines and nuclear receptor in hypoxia.
Hypoxia Plays a Key Role in the Pharmacokinetic Changes of Drugs at High Altitude Current Drug Metabolism, 2018, Vol. 19, No. 11 967

high altitude pulmonary edema or cerebral edema. High altitude CONSENT FOR PUBLICATION
hypoxia not only affects the 140 million people living at high alti- Not applicable.
tude, it also affects an increasing number of people traveling to high
altitude. In recent years, human plateau activity has become in- CONFLICT OF INTEREST
creasingly frequent. Crowds exposed to highland develop acute This article is original, has been written by the stated authors
hypoxia and sometimes even altitude illness. Due to high-altitude who are all aware of its content and approve its submission. It has
emergency situations, and high-altitude military training, and popu- not been published previously, it is not under consideration for
lations living at high altitude, there has been a gradual focus to- publication elsewhere, no conflict of interest exists. The article will
wards understanding drug metabolism at high altitude. High altitude not be published elsewhere in the same form, in any language,
medicine has been developing rapidly, and many research institu- without the written consent of the publisher.
tions of the high altitude disease have been set up. Although the
prevention and treatment of high altitude illness at high altitude has ACKNOWLEDGEMENTS
made great progress, the clinical rational use and the efficacy of
This study was supported by the National Natural Science Fund
drugs has not yet caused enough attention, so it will become very
of China (No. 81760673 and 81460568), and the National Natural
important to establish the subject of high altitude pharmacy and the
Science Fund of Qinghai province (No.2016-ZJ-902). We thank
research institute of drugs in the plateau area.
LetPub (www.letpub.com) for their linguistic assistance during the
How hypoxia influences drug metabolism is a new direction of preparation of this manuscript.
high altitude medicine and drug metabolism research; specifically,
the characteristics and mechanisms of drug-metabolizing enzymes REFERENCES
in these environments, as well as rationale drug use [64, 65]. This [1] Johnson, N.J.; Luks, A.M. High-altitude medicine. Med. Clin.
review primarily focused on studies on acute hypoxia and the North. Am., 2016, 100, 357-369.
CYP450 enzyme system, most of which adopted a low-pressure [2] Shah, N.M.; Hussain, S.; Cooke, M.; O'Hara, J.P.; Mellor, A. Wil-
oxygen cabin to stimulate the plateau environment. These types of derness medicine at high altitude: Recent developments in the field.
studies are not similar enough to the real highland environment and Open Access J. Sports. Med., 2015, 6, 319-328.
cannot give a comprehensive assessment of the effects of hypoxia [3] Zafren, K. Prevention of high altitude illness. Travel. Med. Infect.
on drug PK in those conditions. Thus, although certain findings on Dis., 2014, 12, 29-39.
[4] Netzer, N.; Strohl, K.; Faulhaber, M.; Gatterer, H.; Burtscher, M.
the effects of plateau hypoxia on drug metabolism by the CYP450 Hypoxia-related altitude illnesses. J. Travel. Med., 2013, 20, 247-
enzyme and related drugs have been validated, overall investiga- 255.
tions are still in the exploratory stage. [5] Simon, R.B.; Simon, D.A. Illness at high altitudes. Nursing, 2014,
Preliminary studies have focused on mammals like rats and 44, 36-44.
rabbits, but the drug metabolism of these animals differs from hu- [6] Imray, C.; Wright, A.; Subudhi, A.; Roach, R. Acute mountain
mans. Therefore, studies in humans should be performed to investi- sickness: pathophysiology, prevention, and treatment. Prog. Car-
diovasc. Dis., 2010, 52, 467-484.
gate the effects and mechanism of high-altitude hypoxia. In addi- [7] Pennardt, A. High-altitude pulmonary edema: diagnosis, preven-
tion, studies should be conducted in people native to high altitude, tion, and treatment. Curr. Sports. Med. Rep., 2013, 12, 115-119.
as that 38 million worldwide live in that environment. Furthermore, [8] Li, X.Y.; Gao, F.; Li, Z.Q.; Guan, W.; Feng, W.L.; Ge, R.L. Com-
many ethnic minorities live in plateaus, for example, Tibetan, Hui, parison of the pharmacokinetics of sulfamethoxazole in male Chi-
Tu, Salar, and Mongol nationalities reside in the Qinghai-Tibet nese volunteers at low altitude and acute exposure to high altitude
Plateau. Differences in metabolism among these nationalities vs. subjects living chronically at high altitude: an open-labeled,
should be taken into consideration. Models of acute hypoxia in- controlled, prospective study. Clin. Ther., 2009, 31, 2744-2754.
duced by chemical methods or low-pressure cabin can only stimu- [9] Arancibia, A.; Gai, M.N.; Chávez, J.; Paulos, C.; Pinilla, E.; Gon-
late hypoxia, and not hypothermia, intense radiation, or dry climate, zalez, C.; Villanueva, S.; Ritschel, W.A. Pharmacokinetics of pred-
nisolone in man during acute and chronic exposure to high altitude.
nor is it a real atmosphere. When someone enters the plateau envi- Int. J. Clin. Pharm. Ther., 2005, 43, 85-91.
ronment, it takes a long time to develop hypoxia; this type of re- [10] Li, X.Y. Pharmacokinetics Study of sulfamethoxazole under hy-
striction makes chronic hypoxia difficult to simulate experimen- poxia environment at high altitude. PhD Thesis, Shenyang Pharma-
tally. However, an adaptive process cannot be represented in an ceutical University: Shenyang, 2010.
experimental model of hypoxia. According to the literature reports [11] Powell, J.R.; Vozeh, S.; Hopewell, P.; Costello, J.; Sheiner, L.B.;
and our research, it is generally believed that hypoxia above 2,500 Riegelman, S. Theophylline disposition in acutely ill hospitalized
m altitude will affect the pharmacokinetics of drugs. Therefore, the patient. Am. Rev. Respir. Dis., 1978, 118, 229-238.
real plateau environment (2,500 m) should be used as the ex- [12] Richer, M.; Lam, Y.W. Hypoxia, arterial pH and theophylline
perimental site to overcome limitations of the simulated environ- disposition. Clin. Pharmacokinet., 1993, 25, 283-299.
[13] Letarte, L.; Du Souich, P. Influence of hypercapnia and/or hy-
ment, and factors such as hypoxia, low pressure, strong radiation, poxemia and metabolic acidosis on theophylline kinetics in the
and cold should be taken into account when investigating changes conscious rabbits. Am. Rev. Respir. Dis., 1984, 129, 762-766.
in drug metabolism at high altitude. [14] Du Souich, P. Effect of acute and chronic moderate hypoxia on
Aminophylline, dexamethasone, furosemide, and acetazolamide diltiazem kinetics and metabolism in the dog. Pharmacology.,
have been commonly used for the treatment of acute high-altitude 1993, 47, 378-385.
disease, and their pharmacokinetics in high-altitude environments [15] Zhang, J.H.; Wang, R.; Xie, H.; Yin, Q.; Jia, Z.P.; Li, W.B. Effect
of acute exposure to high altitude on pharmacokinetics of propra-
were previously reported. We should also lay emphasis on cardio- nolol and metoprolol in rats. J. South. Med. Univ., 2014, 34, 1616-
vascular drugs that prevent chronic plateau disease on their meta- 1620.
bolic kinetics study at high altitude in hypoxia, which would offer [16] Li, W.B.; Wang, R.; Xie, H.; Zhang, J.H.; Wu, X.Y.; Jia, Z.P. Ef-
references for plateau rational drug use. The study of activity and fects on pharmacokinetics of propranolol and other factors in rats
expression of drug metabolism enzyme focuses on CYP450 enzyme after acute exposure to high altitude at 4,010 m. Cell. Biochem.
system. In addition, there should be more research on other Biophys., 2015, 72, 27-36.
CYP450 enzymes and phase II metabolism enzymes like NAT2, [17] Zhang, J.H.; Wang, R.; Xie, H.; Yin, Q.; Jia, Z.P.; Li, W.B.; Lu, H.;
UGT, and UST, as current studies seem to mostly focus on cytokine Wang, C.; Sun, Y.H.; Wang, Y.L.; Zhang, X.H.; Hao, Y. Effects of
regulation. A comprehensive understanding is needed of the pathol- aminophylline on the pharmacokinetic parameters at high altitude.
Pharm. J. Chin. PLA., 2014, 30, 125-131.
ogy, cytobiology and molecular biology of the mechanisms under-
lying drug metabolism in high-altitude hypoxic environments.
968 Current Drug Metabolism, 2018, Vol. 19, No. 11 Zhou et al.

[18] Anjana, G.V.; Krishna, K.; Joginder, D. Effect of intermittent hy- drug metabolism in humans. Clin. Pharmacol. Ther., 2002, 71,
pobaric hypoxia on efficacy and clearance of drug. Indian J. Med. 214-220.
Res., 2012, 135, 211-216. [38] Yang, M.; Zhu, J.B.; Li, Y.P.; Fan, X.R.; Yuan, M.; Li, X.Y. The
[19] Shefali, G.; Gaurav, K.K.; Asheesh, G. Hepatic metabolism of effect of high altitude hypoxia on NAT2 activity in rats. J. Qinghai.
ibuprofen in rats under acute hypobaric hypoxia. Exp. Toxicol. Med. Coll., 2013, 34, 79-83.
Pathol., 2013, 65, 751-758. [39] Hao, Y.; Wang, R.; Xie, H.; Sun, Y.H.; Li, W.B.; Jia, Z.P. Influ-
[20] Du, Souich. P. Effect of hypercapnia and/or hypoxemia and meta- ence of plateau environment on enzyme activity of cytochrome-
bolic acidosis on kinetics and concentration of phenytoin in the P450 in drug metabolism and its progress. Chin. J. Pharmacol.
cerebrospinal fluid of conscious rabbits. Neuropharmacology, Toxicol., 2013, 27, 898-902.
1986, 25, 857-862. [40] Du Souich, P.; Fradette, C. The effect and clinical consequences of
[21] Streit, M.; Goggelmann, C.; Dehnert, C.; Burhenne, J.; Riedel, hypoxia on cytochrome P450, membrane carrier proteins activity
K.D.; Menold, E.; Mikus, G.; Bartsch, P.; Haefeli, W.E. Cyto- and expression. Expert. Opin. Drug. Metab. Toxicol., 2011, 7,
chrome P450 enzyme-mediated drug metabolism at exposure to 1083-1100.
acute hypoxia (corresponding to an altitude of 4500 m). Eur. J. [41] Roth, R.A.; Rubin, R.J. Role of blood flow in carbon monoxide and
Clin. Pharmacol., 2005, 61, 39-46. hypoxic hypoxia-induced alterations in hexobarbital metabolism in
[22] Arancibia, A.; Nella, Gai, M.; Paulos, C.; Chávez, J.; Pinilla, E.; rats. Drug. Metab. Dispos., 1976, 4, 460-467.
Angel, N.; Ritschel, W.A. Effects of high altitude exposure on the [42] Du Souich, P.; Saunier, C.; Hartemann, D.; Allam, M. Effect of
pharmacokinetics of furosemide in healthy volunteers. Int. J. Clin. acute and chronic moderate hypoxia on the kinetics of lidocaine
Pharm. Ther., 2004, 42, 314-320. and its metabolites and on regional blood flow. Pulm. Pharmacol.,
[23] Arancibia, A.; Paulos, C.; Chávez, J.; Ritschel, W.A. Pharmacoki- 1992, 5, 9-16.
netics of lithium in healthy volunteers after exposureto high alti- [43] Frandette, C.; Bleau, A.M.; Pichette, V.; Chauret, N.; Du Souich, P.
tude. Int. J. Clin. Harm. Ther., 2003, 41, 200-206. Hypoxia-induced down-regulation of CYP1A1/1A2 and up-
[24] Ritschel, W.A.; Paulos, C.; Arancibia. A.; Pezzani. M.; Agrawal, regulation of CYP3A6 involves serum mediators. Br. J. Pharma-
M.A, Wetzelsberger, K.M.; Lücker, P.W. Pharmacokinetics of col., 2002, 137, 881-891.
meperidine in healthy volunteers after short- and long-term expo- [44] Morgan, E.T. Impact of infectious and inflammatory disease on
sure to high altitude. J. Clin. Pharmacol., 1996, 36, 610-616. cytochrome P450-mediated drug metabolism and pharma-
[25] Li, X.Y.; Liu, Y.N.; Li, Y.P. Pharmacokinetic study of sul- cokinetics. Clin. Pharmacol. Ther., 2009, 85, 434-438.
famethoxazole in healthy Han volunteers living at plain and native [45] Morgan, E.T. Regulation of cytochrome p450 by inflammatory
Han and Tibetan healthy volunteers living at high altitude. Acta. mediators: why and how. Drug. Metab. Dispos., 2001, 29, 207-212.
Pharm. Sin., 2011, 46, 1117-1122. [46] Morgan, E.T.; Li-Masters, T.; Cheng, P.Y. Mechanisms of cyto-
[26] Li, X.Y.; Liu, Y.N.; Wang, X.J.; Zhu, J.B.; Yuan, M.; Li, Y.P.; Li, chrome P450 regulation by inflammatory mediators. Toxicology,
Y.F. Comparison of the pharmacokinetics of sulfamethoxazole in 2002, 181-182, 207-210.
native Han and Tibetan male Chinese volunteers living at high alti- [47] Merry, H.E.; Phelan, P.; Doaks, M.; Zhao, M.Q.; Mulligan, M.S.
tude. Eur. J. Drug. Metab. Pharmacokinet., 2012, 37, 263-269. Functional roles of tumor necrosis factor-alpha and interleukin 1-
[27] Zhang, J.L.; Zhu, J.B.; Yao, X.C.; Duan, Y.B.; Zhou, X.J.; Yang, beta in hypoxia and reoxygenation. Ann. Thorac. Surg., 2015, 99,
M.; Li, X.Y. Pharmacokinetics of lidocaine hydrochloride metabo- 1200-1205.
lized by CYP3A4 in Chinese Han Volunteers living at low altitude [48] Cokic, B.B.; Cokic, V.P.; Suresh, S.; Wirt, S.; Noguchi, C.T. Nitric
and in native Han and Tibetan Chinese volunteers living at high al- oxide and hypoxia stimulate erythropoietin receptor via MAPK
titude. Pharmacology, 2016, 97, 107-113. kinase in endothelial cells. Microvasc. Res., 2014, 92, 34-40.
[28] Preissner, S.C.; Hoffmann, M.F.; Preissner, R.; Preissner, S. Poly- [49] Wenger, R.H. Cellular adaptation to hypoxia: O2-sensing protein
morphic cytochrome P450 enzymes (CYPs) and their role in per- hydroxylases, hypoxia-inducible transcription factors, and O2-
sonalized therapy. PLoS One, 2013, 8, e82562. regulated gene expression. FASEB J., 2002, 16, 1151-1162.
[29] Sevrioukova, I.F.; Poulos, T.L. Understanding the mechanism of [50] Eunjung, L.; Sujin, Y.; Seung, K.L.; Hyunsung, P. Two transactiva-
cytochrome P450 3A4: Recent advances and remaining problems. tion domains of hypoxia-inducible factor-1alpha regulated by the
Dalton Trans., 2013, 42, 3116-3126. MEK-1-p42-p44 MAPK pathway. Mol. Cells, 2002, 14, 9-15.
[30] Proulx, M.; Du Souich, P. Acute moderate hypoxia in conscious [51] Bigham, A.W.; Lee, F.S. Human high-altitude adaptation: forward
rabbits: Effect on hepatic cytochrome P450 and on reactive oxygen genetics meets the HIF pathway. Genes. Dev., 2014, 28, 2189-
species. J. Pharm. Pharmacol., 1995, 47, 392-397. 2204.
[31] Kurdi, J.; Maurice, H.; El-Kadi, A.O.; Ong, H.; Dalkara, S.; Bé- [52] Ji, L.D.; Qiu, Y.Q.; Xu, J.; Irwin, D.M.; Tam, S.C.; Tang, N.L.;
langer, P.M.; Du Souich, P. Effect of hypoxia alone or combined Zhang, Y.P. Genetic adaptation of the hypoxia-inducible factor
with inflammation and 3-methylcholanthrene on hepatic cyto- pathway to oxygen pressure among Eurasian human populations.
chrome P450 in conscious rabbits. Br. J. Pharmacol., 1999, 128, Mol. Biol. Evol., 2012, 29, 3359-3370.
365-373. [53] Park, H. A romatic hydrocarbon nuclear translocator as a common
[32] Fradette, C.; Batonga, J.; Teng, S.; Du Souich, P. Animal models of component for the hypoxia- and dioxin-induced gene expression.
acute moderate hypoxia are associated with a down-regulation of Mol. Cells., 1999, 9, 172-178.
CYP1A1, 1A2, 2B4, 2C5, and 2C16 and up-regulation of CYP3A6 [54] Arnesen, T.; Kong, X.; Evjenth, R.; Lillehaug, J.R. Interaction
and P-glycoprotein in liver. Drug Metab. Dispos., 2007, 35, 765- between HIF-1 alpha (ODD) and hARD1 does not induce acetyla-
771. tion and destabilization of HIF-1 alpha. FEBS Lett., 2005, 579,
[33] Fradette, C.; Du-Souich, P. Hypoxia-inducible factor-1 and activa- 6428-6432.
tor protein-1 modulate the upregulation of CYP3A6 induced by [55] Fisher, T.S.; Etages, S.D.; Hayes, L.; Crimin, K.; Li, B.Y. Analysis
hypoxia. Br. J. Pharmacol., 2003, 140, 1146-1154. of ARD1 function in hypoxia response using retroviral RNA inter-
[34] Li, X.Y.; Wang, X.J.; Li, Y.P.; Yuan, M.; Zhu, J.B.; Su, X.D.; Yao, ference. J. Biol. Chem., 2005, 280, 17749-17757.
X.C.; Fan, X.R.; Duan, Y.B. Effect of exposure to acute and [56] Rahman, M.S.; Thomas, P. Effects of hypoxia exposure on hepatic
chronic high-altitude hypoxia on the activity and expression of cytochrome P450 1A (CYP1A) expression in Atlantic croaker: mo-
CYP1A2, CYP2D6, CYP2C9, CYP2C19 and NAT2 in rats. Phar- lecular mechanisms of CYP1A down-regulation. PLoS One, 2012,
macology., 2014, 93, 76-83. 7, e40825.
[35] Li, X.Y.; Wang, X.J.; Li, Y.P.; Zhu, J.B.; Su, X.D.; Yao, X.C.; Fan, [57] Handschin, C.; Meyer, U.A. Induction of drug metabolism: the role
X.R.; Duan, Y.B. The activity, protein, and mRNA expression of of nuclear receptors. Pharmacol. Rev., 2003, 55, 649-673.
CYP2E1 and CYP3A1 in rats after exposure to acute and chronic [58] Tien, E.S.; Eegishi, M. Nuclear receptors CAR and PXR in the
high altitude hypoxia. High. Alt. Med. Biol., 2014, 15, 491-496. regulation of hepatic metabolism. Xenobiotica, 2006, 36, 1152-
[36] Li, X.Y.; Liu, Y.N.; Yuan, M.; Li, Y.P.; Yang, Y.Z.; Zhu, J.B. The 1163.
effect of high altitude hypoxia on the activity and protein expres- [59] Chen, Y.; Ferguson, S.S.; Negishi, M.; Goldstein, J.A. Induction of
sion of CYP2C9 and CYP2C19. Acta. Pharm. Sin., 2012, 47, 188- human CYP2C9 by rifampicin hyperforin and phenobarbital is me-
193. diated by the pregnane X receptor. J. Pharmacol. Exp. Ther., 2004,
[37] Jürgens, G.; Christensen, H.R.; Brøsen, K.; Sonne, J.; Loft, S.; 308, 495-501.
Olsen, N.V. Acute hypoxia and cytochrome P450-mediated hepatic
Hypoxia Plays a Key Role in the Pharmacokinetic Changes of Drugs at High Altitude Current Drug Metabolism, 2018, Vol. 19, No. 11 969

[60] Hu, B.F.; Bi, H.E.; Huang, M. Advances in the research of preg- volvement in CYP3A4 repression. Eur. J. Cancer., 2009, 45, 2882-
nane X receptor and constitutive and rostane receptor. Acta. Pharm. 2892.
Sin., 2011, 46, 1173-1177. [63] Fradette, C.; Du Souich, P. Effect of hypoxia on cytochrome P450
[61] Vyhlidal, C.A.; Pogan, P.K.; Leeder, J.S. Development and refine- activity and expression. Curr. Drug. Metab., 2004, 5, 257-271.
ment of Pregnane X Receptor (PXR) DNA binding site model us- [64] Lu, H.; Wang, R.; Jia, Z.P.; X, Xiong, J.; Xie, H. Effects of high
ing information theory: insights into PXR-mediated gene regula- altitude exposure on physiology and pharmacokinetics. Curr. Drug.
tion. Biol. Chem., 2004, 279, 46779-46786. Metab., 2016, 17, 559-565.
[62] Legendre, C.; Hori, T.; Loyer, P.; Aninat, C.; Ishida, S.; Glaise, D.; [65] Bailey, D.M. On the significance of altered drug pharmacokinetics
Lucas-Clerc, C.; Boudjema, K.; Guguen-Guillouzo, C.; Corlu, A.; pharmacodynamics at high altitude. High. Alt. Med. Biol., 2017, 18,
Morel, F. Drug-metabolising enzymes are down-regulated by hy- 88-89.
poxia in differentiated human hepatoma Hepa RG cells: HIF-l in-

You might also like