You are on page 1of 17

Arch. Pharm. Res.

(2019) 42:862–878 Online ISSN 1976-3786


https://doi.org/10.1007/s12272-019-01184-3 Print ISSN 0253-6269

REVIEW

Interactions of ginseng with therapeutic drugs


Min‑Koo Choi1 · Im‑Sook Song2

Received: 15 July 2019 / Accepted: 26 August 2019 / Published online: 6 September 2019
© The Pharmaceutical Society of Korea 2019

Abstract Ginseng is the most frequently used herbal Keywords Korean ginseng · Ginsenoside · Herb–drug
medicine for immune system stimulation and as an adju- interaction · Adjuvant chemotherapy
vant with prescribed drugs owing to its numerous pharma-
cologic activities. It is important to investigate the beneficial
effects and interaction of ginseng with therapeutic drugs. Introduction
This review comprehensively discusses drug metabolizing
enzyme- and transporter-mediated ginseng–drug interaction Herbal medicine is increasing being used owing to the effi-
by analyzing in vitro and clinical results with a focus on cacy and low toxicity of traditional plants. Panax ginseng
ginsenoside, a pharmacologically active marker of ginseng. root has been extensively used for more than 2000 years in
Impact of ginseng therapy or ginseng combination therapy East Asian countries because of its ability to restore vital-
on diabetic patients and of ginseng interaction with anti- ity and stimulate the immune system (Lee and Kim 2014;
platelets and anticoagulants were evaluated based on gin- Ru et al. 2015). Ginseng is frequently used owing to its
seng origin and ginsenoside content. Daily administration of therapeutic anti-oxidative, anti-inflammatory, anti-diabetic
Korean red ginseng (0.5–3 g extract; dried ginseng > 60%) properties (Choi 2008) as well as its chemopreventive and
did not cause significant herb–drug interaction with drug adjuvant therapeutic potential (Lu et al. 2009).
metabolizing enzymes and transporters. Among various In the USA, approximately 76% adult population con-
therapeutic drugs administered in combination with gin- sumed herbal supplements in 2017, which increased from
seng, adjuvant chemotherapy, comprising ginseng (1–3 g 64% over the last 9 years (Council for Responsible Nutrition
extract) and anticancer drugs, was effective for reducing 2018). Nearly 25% herbal supplemental users regularly take
cancer-related fatigue and improving the quality of life and prescribed drugs, which have increased the possibility of
emotional scores. Limited information regarding ginseno- herb–drug interaction (Na et al. 2011). Similarly, in seven
side content in each ginseng product and plasma ginsenoside European countries, herbal medicine was used by 9.6–22.7%
concentration among patients necessitates standardization individuals across all age groups, and the concomitant use of
of ginseng product and establishment of pharmacokinetic– therapeutic drugs has been reported in approximately 20%
pharmacodynamic correlation to further understand benefi- children and 60% adults (Jeurissen et al. 2018). Owing to the
cial effects of ginseng–therapeutic drug interactions in future rapid increasing use of herbal medicine and ease of consum-
clinical studies. ing herbal medicine formulations, adverse drug reactions
or herb–drug interactions resulting from co-administration
of therapeutic drugs with herbal medicine have also rap-
* Im‑Sook Song
isssong@knu.ac.kr idly increased (Wu et al. 2016). In China, herbal medicine
1 formulations have accounted for approximately 13.8% and
College of Pharmacy, Dankook University, Cheon‑an 31116,
Republic of Korea 17.3% of the total adverse drug reactions in 2010 and 2013,
2 respectively (Wu et al. 2016). For example, Danshen extract
College of Pharmacy and Research Institute
of Pharmaceutical Sciences, Kyungpook National University, (oral administration, 3 g/day for 10 days) decreased fexofen-
Daegu 41566, Republic of Korea adine plasma exposure via P-glycoprotein (P-gp) induction

13
Vol:.(1234567890)
Interactions of ginseng with therapeutic drugs 863

(Qiu et al. 2014). Co-administration of Schisandrae fruc- this review. Based on the therapeutic benefits of ginseng,
tus extract (oral administration, 600 mg/day for 2 weeks) the pharmacokinetic and pharmacodynamics interactions
increased plasma concentration of talinolol and tacrolimus between ginseng and therapeutic drugs, including antican-
possibly by inhibiting P-gp (Xin et al. 2007; Wu et al. 2016). cer, antiplatelet, anticoagulative, and antidiabetic drugs,
Plasma concentration of cyclosporine, warfarin, oral contra- which can be combined in clinical settings are discussed.
ceptives, theophylline, etc. decreased owing to the induc-
tion of cytochrome P450 (CYP) 3A4, CYP2C9, CYP1A2
enzymes and P-gp (Henderson et al. 2002). Ginsenosides
The most frequently reported herb–drug interaction cases
include herbal medicine modulation by drug-metabolizing Ginsenosides are the major active pharmacological com-
enzymes and transporters and the causative pharmacokinetic ponents of ginseng and unique to ginseng species. Ginse-
effects of co-administered therapeutic drugs that are sub- nosides, also known as steroid-like saponins, are classified
strates for drug-metabolizing enzymes and transporters (Na into two types according to their hydroxylation position on
et al. 2011; Lee et al. 2018), because they are key regula- the core triterpene saponin structure: 20(s)-protopanaxadiol
tors that determine the pharmacokinetics and control the (PPD)-type and 20(s)-protopanaxatriol (PPT)-type ginse-
therapeutic efficacy or adverse reaction of substrate drugs nosides (Won et al. 2019). Ginsenosides found in ginseng
(Seong et al. 2018). Therefore, in vivo results from animal products were subclassified by their sugar moiety at C3,
and clinical studies conducted on ginseng–drug interaction C6, and C20 positions (Fig. 1). These PPD-type ginseno-
and related ginsenoside concentrations will be focused in sides Rb1, Rb2, and Rc undergo further hydrolysis by the

Fig. 1  Structure and metabolic pathway of PPD- and PPT- type ginsenosides. Metabolic pathway represents deglycosylation at C3, C6, or C20
position, catalyzed by beta-glucosidase secreted by the intestinal microflora. PPD: 20(s)-protopanaxadiol; PPT: 20(s)-protopanaxatriol; CK:
compound K, Glc: glucose; Arap: arabopyranose; Araf: arabinofuranose; Rha: rhamnose; Xyl: xylose. The content of ginsenosides in three gin-
seng products (6-year-old Fresh Panax ginseng, red ginseng extract, and white ginseng extract) was given

13
864 M.-K. Choi, I.-S. Song

intestinal microbiota via β-glucosidase; consequently, they rs1189437 TT were associated with increased exposure of
are biotransformed into Rd → F2 or Rg3 → compound K or compound K and decreased exposure of PPD in Chinese
Rh2 → PPD (Fig. 1) (Dong et al. 2017b; Park et al. 2017a). healthy subjects who were orally administered compound
Re and Rg1 belong to PPT-type ginsenosides and are hydro- K (100–400 mg). Genetic variants of pregnane X receptor
lyzed into Rh1 or F1 → PPT (Won et al. 2019) (Fig. 1). (PXR; NR1I2) rs1464602 and rs2472682 were associated
These PPD- and PPT-type ginsenosides and glycosylated with decreased plasma concentrations of compound K. The
metabolites are considered as major active pharmacological results suggested that genetic polymorphisms in MRP4
constituents of ginseng (Ru et al. 2015; Kim et al. 2017b). and PXR also contributed to the inter-subject variability
Therefore, numerous studies have described the immuno- of compound K (Zhou et al. 2019b). Because Compound
logical, antioxidant, anticoagulant, anticancer, antidiabetic, K is reported as a major active component among various
and neuroprotective effects of ginseng and its associated gin- ginsenosides, red ginseng has been fermented using lactic
senosides (Yun et al. 2001; Choi 2008; Gui et al. 2016; Kim acid bacteria to enhance compound K in the ginseng prod-
et al. 2017b; Park et al. 2017b; Lee et al. 2018). uct (Choi et al. 2016). As results, the plasma concentration
Various ginsenosides generally showed low oral bioavail- of compound K in subjects who were orally administered
ability in rats and human. For example, the oral bioavail- 3 g fermented Korean red ginseng (­ Cmax 254.5 ng/mL) as
ability of Rb1 was 1.2–4.4% and that of Rh2 was 4.0–6.4%. substantially higher and less variable than that in subjects
Other ginsenosides including Rg1, Rd, Rh1, and Re showed who received non-fermented Korean red ginseng extract
low oral bioavailability of < 10% (Won et al. 2019). Owing ­(Cmax 8.4–24.8 ng/mL) (Kim 2013; Choi et al. 2016). Taken
to the low oral bioavailability and low intestinal perme- together, limited information regarding plasma ginsenoside
ability of these ginsenosides (Choi et al. 2018), the plasma concentration is available because studies on the pharma-
ginsenoside concentrations are also considerably low. The cokinetics of these ginsenosides garnered lesser interest
maximum plasma concentrations of Rb1, Rb2, Rc, and Rd— than pharmacological in vivo and in vitro studies. However,
major ginsenosides found in rat plasma—were < 10 ng/mL understanding the pharmacokinetics of ginsenosides found
in rats following oral administration of 1.5 g/kg Korean red in human plasma is very important for designing an optimal
ginseng extract (Lee et al. 2018). The maximum plasma con- dose regimen, establishing pharmacokinetic–pharmaco-
centrations of Rb1, Rb2, Rc, and Rd were also < 5 ng/mL dynamic relationship, and understanding the mechanisms
following oral administration of Korean red ginseng extract underlying herb–drug interaction between ginseng products
(dried ginseng > 60%) in healthy Korean subjects (Choi et al. and concomitantly administered therapeutic drugs.
2018). Because these ginsenosides had long elimination Another complication arises from varied ginsenoside
half-lives (> 30 h), the plasma concentrations of Rb1, Rb2, compositions depending on ginseng origin [i.e., Panax
Rc, and Rd after repeated administration for 2 weeks were notoginseng, Panax ginseng (Korean ginseng), and Panax
significantly higher than those after a single oral administra- quinquefolium (American ginseng)] and preparation pro-
tion at the same dose. The accumulation factor, calculated tocols (i.e., fresh ginseng, red ginseng and white ginseng)
by dividing the area under the plasma concentrations (AUC) (Fig. 1). The concentration of some ginsenosides including
value of repeated administration by the AUC value of sin- Rg2, Rg3, Rh1, and Rh2 increases after prolonged steam-
gle administration, was 4.5–6.7 (Choi et al. 2018). Deglyco- ing, whereas that of others (Rb1, Rb2, Rd, Rd, Re, and Rg1)
sylated ginsenosides Rd, Rg3, Rh2, compound K, PPD, and decreases. In Table 1, the characteristics of ginsenosides in
PPT are also found in human plasma following repeated oral Panax notoginseng, Korean red ginseng, Korean ginseng,
administration of Korean red ginseng extract for 2 weeks (Jin and American ginseng are arranged as PPD to PPT ratio,
et al. 2019a). In human plasma, Rd and Rg3 were present at Rb1/Rg1 ratio, and individual contents that are known to
a concentration of 2.2–8.7 ng/mL, whereas Rh2, compound exert antiplatelet and anticoagulant effects. The PPD to PPT
K, PPD, and PPT, which are usually absent in red ginseng ratio is the highest in Korean red ginseng, but it decreases
extract, were present at a concentration of 6.1–81.6 ng/mL. in Korean ginseng and American ginseng and is the lowest
In human plasma, the concentration of compound K was in steamed Panax notoginseng. In Panax ginseng, the pro-
the highest (­ Cmax 81.6 ng/mL), followed by Rb1, Rb2, PPD, portion of PPD-type ginsenosides (2.57–6.67%) is higher
and PPT in that order (12.7 ng/mL, 6.9 ng/mL, 6.1 ng/mL, than that of PPT-type ginsenosides (1.23%). In Korean gin-
and 7.9 ng/mL, respectively) (Jin et al. 2019a). However, the seng, Rb1 and Rb2 are predominant PPD-type ginsenosides
plasma concentrations of compound K have been reported and Rg1 and Re are predominant PPT-type ginsenosides
to show large inter-subject variability due to the variability (Chen et al. 2008). The concentration of ginsenoside Rb1
in the metabolism of compound K from ginseng product (1.51%), Re (0.89%), and Rd (0.77%) in American gin-
mediated by gut microbiota (Kim 2018; Choi et al. 2018; seng is approximately 3, 6, and 5 times higher than that in
Jin et al. 2019a). Genetic variants of multidrug resistance- Korean ginseng and account for > 70% of the total ginseno-
related protein 4 (MRP4; ABCC4) rs1751034 TT and sides. In American ginseng, the Rb1/Rg1 ratio is the highest

13
Interactions of ginseng with therapeutic drugs 865

Table 1  Comparison of ginsenosides among Panax notoginseng, Panax ginseng, and Panax quinquefolium
Composition Steamed Panax Raw Panax Korean Red gin- Panax ginseng Panax quinque- References
notoginseng notoginseng seng extract (Korean ginseng) folium
(American gin-
seng)

Total ginsenoside 50–100 mg/g 50–100 mg/g 30–50 mg/g 20–40 mg/g 40–60 mg/g Lau et al. (2003),
Major ginsenoside R1, Rg1, Rb1, Rd R1, Rg1, Rb1, Rd Rb1, Rb2, Rc, Rd, Rb1, Rb2, Rc, Rb1, Re, Rg1, Rc, Awang and Li,
Rf, Rg1, Rg2, Rg1, Rg1, Re Rd (2008), Lee and
Rg3, Re, Rh1 Kim (2014),
Dong et al.
PPD to PPT ratio 0.9 0.8–2.94 4.2–4.8 2.0–5.4 < 2.0
(2017b), Choi
Rb1/Rg1 1.0 0.56–3.61 0.9–6.6 0.9–2.8 15.3 et al. (2018)
Rb1 15.1 mg/g 26.1 mg/g 4.6–29 mg/g 4.7 mg/g 15.1–49.6 mg/g
Rb2 – – 2.5–3.8 mg/g 0.9 mg/g 0.6 mg/g
Rg1 14.4–18.7 mg/g 19.8–27.6 mg/g 0.7–4.6 mg/g 1.7–5 mg/g 1.3 mg/g

(Table 1), and the ginsenoside concentrations are as follows: or with drug transporters, including organic cation trans-
Rb1 > Re > Rg1 = Rc > Rd. Contrastingly, the Rb1/Rg1 ratio porter (OCT)1, OCT2, organic anion transporter (OAT)1,
is the lowest in Panax notoginseng. Therefore, for achiev- OAT3, organic anion transporting polypeptide (OATP)1B1,
ing better reproducibility and reliable clinical evidence, it OATP1B3, P-gp, and breast cancer resistant protein (BCRP)
is important to standardize ginsenoside concentration using (Table 2).
a pharmacologically effective ginsenoside in in vitro and Contrary to the case of ginseng product, individual ginse-
in vivo studies with ginseng product. nosides modulate drug-metabolizing enzymes or transport-
ers. For example, ginsenoside Rb1 significantly inhibited
CYP2C9, UGT1A9, OATP1B1, and OATP1B3 with ­IC50
Effect of ginseng on drug‑metabolizing enzyme values of 2.4 µM, 21.3 µM, 33.2 µM, and 4.8 µM, respec-
and transporters tively, whereas other CYP and UGT enzymes as well as
other transporters remained unaffected (Table 2). In hepato-
In vitro study cyte induction experiments, Rb1 either did not induce or
inhibited mRNA expression of CYP and UGT enzymes as
Drug-metabolizing enzymes and transporters play cru- well as of P-gp and BCRP transporters (Table 2).
cial roles in the pharmacokinetics, safety, and efficacy of Ginsenoside Rg3 inhibited UGT1A3, UGT1A9, and
therapeutic drugs by modulating the absorption, distribu- UGT2B7 with I­C 50 values of 20.9 µM, 15.1 µM, and
tion, metabolism, and excretion (ADME) properties of their 23.1 µM, respectively. Ginsenoside Rh2 inhibited UGT1A3
substrate drugs. For example, bioavailability, metabolism, with ­IC50 value of 37.9 µM (Kim et al. 2016a). ­IC50 values
and elimination of the major active components should be for individual ginsenosides are relatively high considering
investigated in herbal medicine. The possible interactions the plasma ginsenoside concentrations in humans; therefore,
of herbal medicines with co-administered drugs should be the possibility of ginsenoside–drug interaction is remote.
monitored and classified according to the pharmacokinetic However, OATP1B1 and OATP1B3 transporters were
fold change in the affected therapeutic drugs based on the strongly inhibited by ginsenosides. Ginsenosides Rb1, Rc,
values of I­ C50 or E
­ C50 (i.e., half maximal concentration of and Rd (PPD-type ginsenoside) inhibited OATP1B1 and
inhibition or induction of enzyme activity or expression) of OATP1B3 with ­IC50 values of 0.2–4.6 µM. Rg1, and ginse-
Panax ginseng product (Seong et al. 2018). noside Re (PPT-type ginsenoside) also inhibited OATP1B1
Seong et al. 2018 reported in vitro inhibitory and induc- and OATP1B3 with I­ C50 values of 39.4–133 µM (Jiang et al.
ible effects of red ginseng on drug-metabolizing enzymes 2015). Although the clinical herb–drug interaction of gin-
and transporters, to evaluate the potential of ginseng product seng or ginsenoside associated with OATP1B1 or OATP1B3
as a perpetrator of pharmacokinetic drug interactions, and has not yet been investigated, caution should be taken with
the results are shown in Table 2. In vitro drug interaction long-circulating PPD-type ginsenoside following the admin-
studies suggest that ginseng product does not cause clini- istration of high-dose Korean red ginseng or PPD-type gin-
cally relevant drug–drug interaction with drug-metabolizing senoside. Regarding this issue, a case of drug-induced liver
enzymes, including CYP1A2, CYP2B6, CYP2C8, CYP2C9, injury (DILI) in patient (82-year-old, male) who took ator-
CYP2C19, CYP2D6, CYP3A4, UDP-glucuronosyltrans- vastatin (80 mg), atenolol (50 mg), and aspirin (100 mg)
ferase (UGT)1A1, UGT1A4, UGT1A9, and UGT2B7, after concomitant ginseng intake was reported based on

13
866

Table 2  Summaries of in vitro system to evaluate the effect of Panax ginseng on the activities of drug-metabolizing enzyme and transporters
System Substrate Metabolite IC50 (µg/mL) (Seong et al. 2018)

13
Fresh ginseng Red ginseng extract White ginseng extract Rb1

CYPs
CYP1A2 Human liver micro- Phenacetin acetaminophen NI NI NI NI
CYP2A6 somes Coumarin OH-coumarin NI NI NI NI
CYP2B6 Bupropion OH-bupropion 112 NI NI NI
CYP2C8 Paclitaxel OH-paclitaxel NI NI NI NI
CYP2C9 Diclofenac OH-diclofenac NI NI NI 2.4
CYP2C19 Omeprazole OH-omeprazole NI NI NI NI
CYP2D6 Dextromethorphan Dextrorphan NI NI NI NI
CYP3A4 Midazolam OH-midazolam NI NI NI NI
UGTs
UGT1A1 Human liver micro- SN-38 SN-38-glucuronide NI NI NI NI
UGT1A3 somes Chenodeoxycholic Chenodeoxycholate- NI NI NI NI
acid glucuronide
UGT1A4 Trifluoperazine Trifluoperazine- NI NI NI NI
glucuronide
UGT1A6 N-acetylserotonin N-acetylserotonin- NI NI NI NI
glucuronide
UGT1A9 Mycophenolic acid Mycophenolic acid- NI NI NI 21.3
glucuronide
UGT2B7 Naloxone Naloxone-glucuronide NI NI NI NI
Transporters
OCT1 Overexpressed system N-methyl-4-phe- NI NI NI NI
in HEK293 cells nylpyridinium
OCT2 N-methyl-4-phe- NI NI NI NI
nylpyridinium
OAT1 p-amino hippuric NI NI NI NI
acid
OAT3 Estrone-3-sulfate NI NI NI NI
OATP1B1 Estrone-3-sulfate NI NI NI 33.2
OATP1B3 Estradiol-17-beta- 104 62 104 4.8
d-glucuronide

BCRP Overexpressed system Estrone-3-sulfate NI NI NI NI


P-gp in LLC-PK1 cells Digoxine NI NI NI NI

IC50: Half maximal inhibitory concentration; NI: No significant inhibition


CYP cytochrome P450, UGT​ UDP-glucuronosyltransferase, OCT organic cation transporter, OAT organic anion transporter, OATP organic anion transporting polypeptide, P-gp P-glycoprotein,
BCRP breast cancer resistance protein
M.-K. Choi, I.-S. Song
Interactions of ginseng with therapeutic drugs 867

the thorough medication history and the close examination. in rats, which could be explained by reduced fexofenadine
After the cessation of both atorvastatin and ginseng product, absorption caused via induction of intestinal P-gp expres-
the patient’s symptoms was resolved within 2 months (Laube sion (Zhang et al. 2009). However, repeated red ginseng
and Liu 2019). The inhibition of CYP3A4 and/or OATP1B1 extract treatment did not modulate Bsep and P-gp expres-
activities by ginseng and causative impaired elimination of sion in the liver but decreased Mrp2 mRNA and protein
atorvastatin could be suspected for the patient’s hepatotoxic- expression (Lee et al. 2018). Intestinal Oct1 expression
ity (Laube and Liu 2019). increased but hepatic Oct1 expression decreased in rats
owing to repeated administration of Korean red ginseng
In vivo animal study extract (1.5 g/kg for 7 days). Although intestinal Cyp3a
decreased owing to repeated administration of Korean
In in vivo pharmacokinetic herb–drug interaction study, a red ginseng extract (1.5 g/kg for 7 days), hepatic Cyp3a
single dose of Korean red ginseng extract was administered remained unaffected (Jin et al. 2019b). It is speculated
to experimental animals to investigate whether Korean red that ginsenoside concentrations in the intestine and liver
ginseng could directly affect the pharmacokinetic properties were considerably different because of its low bioavail-
of substrate drugs by inhibiting the critical pharmacokinetic ability; thus, different ginsenoside concentrations could
pathway including drug-metabolizing enzymes and trans- differentially modulate the metabolizing enzymes and
porters. Contrastingly, repeated administration of Korean transporters depending on the tissue type. It could be also
red ginseng extract was performed to investigate whether speculated that the increase in mRNA levels of Hnf-1α in
Korean red ginseng extract treatment could regulate the the liver resulting from oral administration of red ginseng
expression of drug-metabolizing enzymes and transporters extract at high doses (30 mg/kg for 10 days) (Bogacz et al.
(Jin et al. 2019b). 2016) differentially regulates drug-metabolizing enzymes
Jo and Lee (Jo and Lee 2017) reported no herb–drug and transporters depending on the tissue type, which in
interaction between single oral dose of Korean red gin- turn complicates in vivo ginseng–drug interaction.
seng extract (0.5–2.0 g/kg) and 5 Cyp enzymes (i.e.,
Cyp1a, Cyp2b, Cyp2c, Cyp2d, and Cyp3a). Repeated oral In vivo human study
administration of Korean red ginseng extract (0.5 g/kg
for 2 weeks; equivalent to 4.05 mg ginsenoside/kg) did Several studies performed to investigate ginseng–drug inter-
not alter the metabolic activity of Cyp1a, Cyp2b, Cyp2c, actions and results are briefly summarized in Table 3. Gurley
Cyp2d, and Cyp3a in the mouse liver (Kim et al. 2012). et al. (Gurley et al. 2002, 2005) investigated metabolic activity
Another study revealed that oral administration of etha- changes in CYP1A2, CYP2E1, CYP3A4, and CYP2D6 fol-
nol extract of ginseng (30 mg/kg for 10 days; equivalent lowing oral administration of 0.5 g Korean ginseng powder
to 8.13 mg ginsenoside/kg) to rats increased the mRNA twice daily for 4 weeks in young and elderly heathy subjects.
expression of Cyp2d1 and Cyp3a2 in rat liver (Bogacz No significant change was reported in CYP-mediated metabo-
et al. 2016). The discrepancy between the two studies lism in both populations.
can be explained from different ginsenoside concentra- In a study conducted by Malati et al. (2012), Korean gin-
tions and compositions of the ginseng product used. seng (0.5 g capsule twice daily for 28 days) induced CYP3A
This treatment also increased the mRNA expression of activity following oral administration of 8 mg midazolam,
hepatocyte nuclear factor (Hnf)1α but not of Hnf4α in and the geometric least square mean ratio for AUC (GMAR)
the liver (Bogacz et al. 2016), which is one of the master (90% confidence intervals) was 0.66 (0.55–0.78). Of the six
transcription factor that differentially regulates numerous studies enlisted in Table 3, this is the only significant result
drug-metabolizing enzymes and transporters (Shih et al. on CYP3A4 induction following repeated administration of
2001; Maher et al. 2006; Martovetsky et al. 2013). Korean ginseng.
Repeated administration of ginseng extract report- Kim et al. (2016b) investigated the effects of Korean red
edly regulates the expression of drug transporters in a ginseng extract on the activities of CYP enzymes in 15 healthy
tissue-specific manner. The expression of Oat1 and Oat3 volunteers, by administering them with a modified Inje cock-
in the kidney and the expression of P-gp in the liver was tail (Ryu et al. 2007). Korean red ginseng extract (64% dried
increased by the repeated administration of Korean red ginseng, once daily for 2 weeks) weakly inhibited CYP2C9
ginseng extract in a dose dependent manner (30–300 mg/ and CYP3A4 and weakly induced CYP2D6, with no clini-
kg for 2 weeks; equivalent to 13 mg ginsenoside/kg) in cally significant interactions observed between red ginseng and
mice, which was accompanied with the dose dependent CYP enzymes (Kim et al. 2016b) (Table 3). In another study
decrease of fexofenadine AUC (Kim et al. 2018). Repeated by Kim et al. (2016c) involving 2-week treatment with concen-
administration of Ginseng radix extract (150 mg/kg/day) trated fermented red ginseng liquid (> 3% dried ginseng, once
for 2 weeks reduced fexofenadine bioavailability by 16.1% daily for 2 weeks), no significantly different drug interactions

13
Table 3  Clinical studies to evaluate the effect of ginseng on the activities of drug-metabolizing enzyme and transporters
868

Subject character- Age Ginseng dose regimen Phenotype assessment Results References
istics

13
Healthy, young 25.0 ± 3.9 Panax ginseng 0.5 g, tid/day (75 mg ginse- CYP3A4 ([OH-midazolam]/[midazolam] at No significant difference in CYP3A4, Gurley et al. (2002)
12 (M6:F6) noside/day) for 4 weeks 1-h plasma) CYP1A2, CYP2E1, and CYP2D6
CYP1A2 ([paraxanthine]/[caffeine] at 6-h between control and ginseng groups
plasma) Panax ginseng had little effect on CYP-
CYP2E1([OH-chlorozoxazone]/[chlorozo- mediated metabolism
xazone] at 2-h plasma)
CYP2D6([OH-debrisoquin]/[debriso-
quin + OH-debrisoquin] for 8-h urine
recovery)
Healthy, elderly 67.0 ± 5.2 Panax ginseng 0.5 g, tid/day (75 mg ginse- CYP3A4 ([OH-midazolam]/[midazolam] at No significant difference in CYP3A4, Gurley et al. (2005)
12 (M6:F6) noside/day) for 4 weeks 1-h plasma) CYP1A2, CYP2E1, and CYP2D6
CYP1A2 ([paraxanthine]/[caffeine] at 6-h between control and ginseng groups
plasma) Panax ginseng had little effect on CYP-
CYP2E1([OH-chlorozoxazone]/[chlorozo- mediated metabolism
xazone] at 2-h plasma)
CYP2D6([OH-debrisoquin]/[debriso-
quin + OH-debrisoquin] for 8-h urine
recovery)
a
Healthy 32.3 ± 6.7 Panax ginseng 0.5 g, bid/day (50 mg gin- CYP3A4 (AUC of midazolam) Significant increase in CYP3A4 activity in Malati et al. (2012)
12 (M8:F4) senoside/day) for 4 weeks P-gp (AUC of fexofenadine) ginseng group
No significant difference in P-gp activity in
ginseng group
b
Healthy 24.5 ± 1.9 Korean red ginseng extract (100 mg ginse- CYP1A2 (GMMR of paraxanthine/caf- Korean red ginseng weakly inhibited Kim et al. (2016b)
14 (M14:F0) noside/day) for 2 weeks feine) CYP2C9 and CYP3A4 activities and
CYP2C9 (GMMR of EXP3174/losartan) weakly induced CYP2D6 activity
CYP2C19 (GMMR of OH-omeprazole/ Korean red ginseng had little effect on
omeprazole) CYP1A2, CYP2C19, and P-gp function
CYP2D6 (GMMR of dextrorphan/dex-
tromethorphan)
CYP3A4 (GMMR of OH-midazolam/
midazolam)
P-gp (GMAR of fexofenadine)
Healthy 25.6 ± 2.6 Fermented red ginseng extract (ginsenoside CYP1A2 (GMAR of paraxanthine and Korean red ginseng weakly inhibited Kim et al. (2016c)
15 (M15:F0) content not specified) for 2 weeks caffeine) CYP1A2, CYP2C9, and CYP3A4 activi-
CYP2C9 (GMAR of EXP3174 and losar- ties.
tan) Korean red ginseng had little effect on
CYP2C19 (GMAR of OH-omeprazole and CYP2C19, CYP2D6, and P-gp function
omeprazole)
CYP2D6 GMAR of dextrorphan and dex-
tromethorphan)
CYP3A4 (GMAR of OH-midazolam and
midazolam)
P-gp (GMAR of fexofenadine)
M.-K. Choi, I.-S. Song
Interactions of ginseng with therapeutic drugs 869

were reported between fermented red ginseng and CYP probe

Daily intake of Korean red ginseng extract comprises 100 mg of total ginsenosides including 29.02 mg Rb1, 4.63 mg Rb2, 15.68 mg Rc, 8.58 mg Re, 4.59 mg Rg1, 15.04 mg Rh1, and

M male, F female, tid ter in die, three times a day; bid bis in die, twice a day, GMMR Geometric least square mean metabolic ratio, GMAR Geometric least square mean AUC ratio, CYP
Daily intake of Korean red ginseng extract comprises 85 mg of total ginsenosides including 23.0 mg Rb1, 11.4 mg Rb2, 13.0 mg Rc, 6.6 mg Rd, 6.2 mg Re, 2.8 mg Rg1, 8.0 mg Rh1, and
Seong et al. (2018)
drugs, with a weak inhibition of CYP1A2, CYP2C9, and
CYP3A4 (Table 3). Seong et al. (2018) also reported no clini-
References

cally significant inhibitory effects between Korea red ginseng


and CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4,
and OATP1B1 probe substrates following single or multiple
administration of Korean red ginseng extract (> 60% dried gin-
seng had no clinically significant effect on
CYP2C9, CYP2C19, CYP2D6, CYP3A4,

CYP1A2, CYP2C9, CYP2C19, CYP2D6,


Repeated administration of Korean red gin-
seng, once daily for 2 weeks) (Table 3). Because these clini-
ginseng had little effect on CYP1A2,

cal studies conducted using Korean red ginseng product had


Single administration of Korean red

CYP3A4, and OATP1B1 function


similar ginsenoside content and origin, clinical results were
consistent. In summary, Korean red ginseng did not exhibit
significant pharmacokinetic herb–drug interaction with drug-
and OATP1B1 function

metabolizing enzymes and transporters, regardless of a single


or repeated administration. This could be attributed to the low
absorption and low plasma concentration of ginsenosides, con-
sistent with the in vitro prediction in a previous study (Choi
et al. 2018; Seong et al. 2018).
Results

Daily intake of Panax ginseng root powder comprises 50 mg of total ginsenosides including 14.78 mg Rb1 and 13.6 mg Rb2

Effect of ginseng on therapeutic drugs


CYP2C19 (GMAR of OH-omeprazole and
CYP2C9 (GMAR of EXP3174 and losar-

CYP3A4 (GMAR of OH-midazolam and


CYP1A2 (GMAR of paraxanthine and

CYP2D6 (GMAR of dextrorphan and

Anticancer drugs
OATP1B1 (GMAR of pitavastatin)

Numerous in vitro and in vivo animal studies have proved


the effect of ginseng or ginsenosides on increased cytotox-
Phenotype assessment

icity of chemotherapeutic agents including 5-fluoroura-


dextromethorphan)

cil, irinotecan, mitomycin C, docetaxel, and cisplatin at a


omeprazole)

concentration range of 0.1–300 µg/mL (Chen et al. 2014).


midazolam)
caffeine)

Repeated oral dose of PPD (70 mg/kg) for 6 weeks increased


the plasma exposure of calcitriol in prostate tumor bearing
tan)

mice, which is attributed to the decreased Cyp3a activity that


are responsible for the metabolism of calcitriol. The results
Korean red ginseng extract (85 mg ginse-

suggested enhanced efficacy of calcitriol with a co-treatment


of PPD (Ben-Eltriki et al. 2019).
A critical concern in the above mentioned in vitro and
in vivo animal studies was the relatively high concentra-
Single dose and for 2 weeks

tion of ginseng extract and ginsenoside because this con-


centration is not likely achievable in humans owing to the
Ginseng dose regimen

maximum plasma ginsenoside concentration reportedly


being < 100 ng/mL (Chen et al. 2008; Choi et al. 2018).
noside/day)

Therefore, it is critical to identify individual ginsenosides


that shows anti-tumor efficacy at a low dose. Ginsenoside
Rg3 is effective against various human cancer cells, and
cytochrome P450, P-gp P-glycoprotein

when used at doses of 3–20 mg/kg in combination with


c

anticancer drugs, including cyclophosphamide and gem-


citabine, it improves survival rate in tumor-bearing animal
24.9
Age

models (Chen et al. 2014). Rg3 suppresses tumor growth and


inhibits tumor angiogenesis by inhibiting vascular endothe-
Table 3  (continued)

lial growth factor-dependent pathway (Xu et al. 2016). Rg3


Subject character-

also induces apoptosis and cell cycle arrest at G0/G1 phase


14.45 mg Rg3
15 (M15, F0)

14.1 mg Rg3

and decreases drug resistance by inactivating nuclear fac-


Healthy

tor kappa light chain enhancer of activated B cells (NF-kB)


istics

(Chen et al. 2014). These findings were substantial clinical


b
a

13
870 M.-K. Choi, I.-S. Song

evidence for Rg3 efficacy and the basis for the development life (Table 4). High dose of Korean ginseng and American
of ginsenoside Rg3 capsule (20 mg; Shenyi®, China). Rg3 ginseng (over 1000 mg/day for > 4 weeks) in combination
(20 mg twice daily for 30 days) administered in combina- with conventional chemotherapy significantly improves
tion with gemcitabine and cisplatin in advanced esophageal CRF without causing any critical adverse event in patients
cancer patients increased 1-year survival rate and reduced with different cancer types and stages (Table 4). Two clini-
drug-associated adverse events; this adjuvant triple therapy cal studies involving administration of Korean red ginseng
substantially improved quality of life as opposed to chemo- and fermented red ginseng at 3000 mg/day in combination
therapy using gemcitabine and cisplatin alone (Chen et al. with platinum-based chemotherapy also improved CRF
2014). Meta-analysis of 20 clinical studies involving admin- score (Table 4). The patients showed significantly improved
istration of Rg3 (20 mg twice daily 6–24 weeks) combined emotional symptoms and quality of life scores and reduced
with chemotherapy for non-small cell lung cancer patients anticancer drug-related toxicity. During the study period,
indicated that Rg3 enhances response rate, prolongs overall high-dose ginseng administration was tolerated by cancer
survival rate, reduces treatment-related toxicity, and pro- patients. However, in both cases, survival rates and tumor
motes and improves quality of life (Xu et al. 2016). markers did not significantly change after administration of
Ginseng product is also frequently administered as adjuvant therapy with a high dose of ginseng (Kim et al.
a health supplement along with cancer chemotherapy. 2017a; Jiang et al. 2017). An interesting results on the inter-
Because many anticancer drugs are substrates for CYP3A4 action between cisplatin and ginsenoside Rb1 have been
and P-gp, ginseng–anticancer drug interaction has been reported in A549 lung cancer bearing mice (Zhou et al.
focused on CYP3A4 and P-gp. No significant in vivo phar- 2019a). The intervention of cisplatin (2 mg/kg, intraperito-
macokinetic interactions between ginseng and CYP3A4 and neal injection) significantly decreased the plasma exposure
P-gp function have been reported, except for the findings of Rd, Rg3, and F2, metabolites of Rb1, but did not affect
reported by Gurley et al. (Table 3). However, two contro- the plasma concentrations of Rb1, suggesting the inhibi-
versial ginseng–drug interaction cases with regard to cancer tion of gut metabolism from Rb1 to Rd, Rg3, and F2 from
therapy have been reported (Table 4). Both cases deduced Rb1 by cisplatin treatment (Zhou et al. 2019a). The results
that decrease in CYP3A4 activity or induction of CYP3A4 were supported by the decreased deglycosylation of Rb1 but
can be attributed to imatinib-associated liver toxicity or increased intestinal permeability of Rb1 by cisplatin (Zhou
gefitinib resistance, respectively (Hwang et al. 2008; Bilgi et al. 2019a). Taken together, the beneficial anti-proliferative
et al. 2010). However, the underlying mechanisms related to effect of ginseng needs further validation in cancer treatment
CYP3A4 activity and the associated controversies need to considering the effect of platinum-based chemotherapy on
be carefully validated. the inhibited gut metabolism, narrowed intestinal absorptive
In order to improve the clinical interpretation of the area, increased efflux ratio of ginsenoside, and as enhanced
in vivo pharmacokinetic interactions between herbal medi- intestinal permeability (Zhou et al. 2019a).
cines and chemotherapy, a novel algorithm-assisted infor-
mation system has been developed and visualized by the Antiplatelet drugs
grading colors according to the risk categories (Ziemann
et al. 2019). In this system, cyclosporine, cyclophospha- Lau et al. (2009) reported the different antiplatelet effects
mide, docetaxel, imatinib, irinotecan, and tamoxifen were of Panax notoginseng, Korean ginseng, and American gin-
indicated as possible interaction with ginseng mediated by seng. Platelet inhibition percentage following treatment with
CYP3A4 and/or P-gp (Ziemann et al. 2019). However, only these ginseng products (3 mg/mL) was compared with that
3 clinical trials and 1 case report on pharmacokinetic inter- following treatment with 200 µM aspirin (positive control);
actions between ginseng and anticancer drugs were included similarly, steamed Panax notoginseng showed an equivalent
in this algorithms. Prediction of pharmacokinetic interac- effect with aspirin, and the effect decreased in the order of
tion potential from in vitro cell system could be restricted raw Panax notoginseng, Korean red ginseng, Korean gin-
because of lack of information on the plasma concentration seng, and American ginseng. Ginsenosides Rg1 and Rg2
of active components of ginseng, ginsenosides (Ziemann were reported as the main antiplatelet components (Teng
et al. 2019). Further studies for determining plasma ginse- et al. 1989; Kuo et al. 1990).
noside concentrations and the used of standardized ginseng Ginsenosides Rg1 and Rg3 inhibited thrombin-induced
product may be useful for the extrapolation of clinical gin- human platelet aggregation in a dose-dependent manner
seng–drug interactions. (Kwon 2018). Difference in antiplatelet effects depend on
Clinical studies on pharmacodynamic interactions the origin of ginseng can be attributed to the different gin-
between ginseng and anticancer drugs have been focused on senoside contents of ginseng since ratios of Rb1/Rg1 and
the effect of ginseng in adjuvant chemotherapy for reducing PPD/PPT are the lowest in Panax notoginseng but the high-
cancer-related fatigue (CRF) and for improving quality of est in American ginseng (Table 1). The concentrations of

13
Table 4  Clinical studies evaluating the effect of ginseng on anticancer drugs
Study Subject characteristics Age Ginseng dose regimen Phenotype assessment Results References
(+Combination drugs)

Pharmacokinetics interac- Case report 36-year-old (F) Complementary herbal Chest CT scan Initial 9 weeks of gefitinib Hwang et al. (2008)
tion: possibly via CYP3A Stage IV lung adenocarci- medicines including along with ginseng, pri-
induction noma ginseng gefitinib, 250 mg/ mary mass and metastasis
day PO increased 4 weeks of gefi-
tinib with discontinuation
of complementary herbal
medicines, primary mass
and metastasis markedly
decreased
Continuation of gefitinib for
30 weeks, primary mass
Interactions of ginseng with therapeutic drugs

and metastasis further


decreased
Pharmacokinetics interac- Case report 26-year-old (M) Energy drink containing Liver toxicity 7 years of imatinib mainte- Bilgi et al. (2010)
tion: possibly via CYP3A Chronic myelogenous ginseng nance and follow-up
or P-gp inhibition leukemia Imatinib, 400 mg/day PO Imatinib-associated liver
toxicity was induced dur-
ing 3 months of imatinib
therapy along with ginseng
After treatment of liver
toxicity, imatinib was
restarted at the same dose
without recurrence of liver
toxicity
Adjuvant chemotherapeu- 282 American ginseng, 750, Multidimentional fatigue No significant improvement Arring et al. (2018)
tic agent: cancer-related (M96:F186) 1000, and 2000 mg for symptom inventory at a dose of 750 mg
fatigue (CRF) 8 weeks Decreasing CRF at doses of
1000 and 2000 mg
364 American ginseng, 2000 mg Multidimentional fatigue Improved fatigue score at Barton et al. (2013)
(M80:F284) for 8 weeks symptom inventory ginseng dose of 2000 mg
40 Panax ginseng, 1252.5 mg Visual analogue scale of Significantly improved Jeong et al. (2010)
(M16:F24) for 2 weeks global fatigue fatigue score
30 Panax ginseng, 800 mg for Functional assessment of Significantly improved Yennu et al. (2016)
(M15:F15) 4 weeks chronic illness therapy fatigue score
fatigue
15 52.4 ± 9.9 Methylphenidate 10–40 mg/ Change in fatigue score in Significant reduction in Chang et al. (2018)
(M7:F8) day + American gin- numerical rating scale fatigue
seng 2000 mg/day for
30.5 ± 7.8 days

13
871
Table 4  (continued)
872

Study Subject characteristics Age Ginseng dose regimen Phenotype assessment Results References
(+Combination drugs)

13
Adjuvant chemotherapeutic Epithelial ovarian cancer 55.9 ± 12.1 6 cycles of taxane and Genotoxicity-binucleated No difference in binucleated Kim et al. (2017a)
agent: genotoxicity and patients platinum-based chemother- cells index and micronu- cell index after ginseng
HRQL 30 apy + 1Korean red ginseng clei yield treatment
3000 mg/day for 3 months HRQL–Functional scale, Significant reduction in
symptom scale micronuclei yield after
Brief fatigue inventory ginseng treatment
Anxiety and depression Significantly improved
scale HRQL score
Significantly improved
fatigue score
Significantly improved anxi-
ety and depression score
Non-small cell lung cancer 58.6 ± 19.3 Gemcitabine 1 g/m2 Chemotherapy toxicity Significantly decreased leu- Jiang et al. (2017)
patients and cisplatin 25 mg/ evaluation kopenia, thrombocytope-
60 (M42:F18) m2 chemotherapy every Comparison of tumor nia, nausea, and vomiting
3 weeks + fermented red marker No difference in tumor
ginseng 3000 mg/day for Anxiety and depression marker after ginseng treat-
60 days scale ment
Functional assessment of Significantly improved anxi-
chronic illness therapy ety and depression score
fatigue Significantly improved
fatigue score
a
Korean red ginseng 1 g contains ginsenoside Rb1 (5.61 mg/g), Rb2 (2.03 mg/g), Rc (2.20 mg/g), Rd (0.39 mg/g), Re (1.88 mg/g), Rf (0.89 mg/g), Rg1 (3.06 mg/g), Rg2 (0.15 mg/g), Rg3
(0.17 mg/g), and Rh1 (0.30 mg/g)
M male, F female, HRQL health-related quality of life, CT computed tomography
M.-K. Choi, I.-S. Song
Interactions of ginseng with therapeutic drugs 873

individual components are more important than the total inappropriate interventions could result in unexpected clini-
ginsenoside content in ginseng product. cal outcomes (Leite et al. 2016; Choi et al. 2017). Some
The importance of Panax notoginseng ginsenosides (R1, studies produced superficial results, and other articles which
Rg1, Rb1, Rd, and Re) in cardiovascular diseases, and the reported cases of interactions between warfarin and ginseng
interaction between aspirin and Panax notoginseng has did not provide relevant clinical outcome-related informa-
been investigated in rats. Because of a very short half-life tion or quantify the clinical significance (Leite et al. 2016;
of aspirin and rapid transformation of salicylate from aspi- Agbabiaka et al. 2017).
rin, Tian et al. (2017) compared salicylate concentrations Recently, interactions between ginseng and warfarin
following co-administration of aspirin and Panax notogin- have been reported to decrease warfarin efficacy owing to
seng (30.25 mg/kg as total ginsenosides) with that following its pharmacokinetic alterations. The concomitant use of con-
administration with aspirin alone. Plasma concentration of centrated ginseng extract interrupted the warfarin-caused
salicylate increased by twofolds following co-administration increase in the international normalized ratio (INR) in a
with Panax notoginseng. Intestinal permeability of aspirin time-dependent and dose-dependent manner (Dong et al.
and salicylate was significantly increased by the presence 2017a). A well-established K-carrageenan-induced rat tail
of Panax notoginseng ginsenosides in Caco-2 cells, sug- thrombosis model demonstrated that ginsenoside caused
gesting that pharmacokinetic interactions between aspirin dose-dependent antagonism of the anti-coagulation effect
and Panax notoginseng could occur during absorption (Tian of warfarin after repeated co-administration of concentrated
et al. 2017). Aspirin increases plasma concentrations and ginseng extract (30, 100, 300 mg/kg for 3 weeks) and war-
intestinal permeability of ginsenosides R1, Rg1, Rb1, Re, farin; consequently, decreased plasma concentrations of
and Rd when concomitantly administered, and the mecha- warfarin and increased warfarin metabolism in turn reduced
nisms underlying increased absorption were that Panax warfarin efficacy owing to CYP3A4 and CYP2C9 upregula-
notoginseng ginsenosides increase membrane fluidity and tion (Dong et al. 2017a).
that aspirin and salicylate open up the tight junction of cells Several clinical studies also focus on this issue (Table 5).
(Tian et al. 2018). These results suggest likely clinical phar- American ginseng exhibited herb–drug interaction with
macodynamics and pharmacokinetic interactions between warfarin, causing a reduction in the anti-coagulative effect
Panax notoginseng and aspirin, but associated clinical evi- exerted by warfarin (Yuan et al. 2004). However, other three
dence remains unavailable. studies (Jiang et al. 2004; Lee et al. 2008, 2009) involving
Korean ginseng had little effect on warfarin pharmacoki-
Anticoagulant drugs netics and efficacy, despite the study subjects being admin-
istered a high ginsenoside dose (100 mg/day for 6 weeks)
Warfarin is the most common oral anticoagulant used for (Table 5). Therefore, similar to the case of aspirin, the ginse-
preventing thromboembolic disorders. It has a narrow noside composition and concentration seems to be important
range between therapeutic and toxic doses, suggesting that and should be studied further.

Table 5  Clinical studies investigating the herb–drug interaction between warfarin and ginseng
Subject characteristics Age Ginseng dose Duration Results References

Healthy 30.2 ± 7.2 Warfarin 5 mg 4 weeks American ginseng reduced the Yuan et al. (2004)
20 (M9:F11) American ginseng 0.5 ga anti-coagulation effect of
warfarin
Healthy 20–40 Warfarin 25 mg 2 weeks Korean ginseng had little effect Jiang et al. (2004)
12 (M12:F0) Korean ginseng 0.5 gb on warfarin INR and warfarin
metabolism
Cardiac valve replacement < 50: 8 Warfarin 40.60 ± 14.53 mg/ 6 weeks Korean red ginseng had little Lee et al. (2009)
patients 50–60: 9 week effect on the anti-coagulation
25 (M4:F21) > 60: 8 Korean red ginseng 1.0 gc effect of warfarin
Ischemic stroke patients 62.75 ± 7.71 Warfarin 2–5 mg 2 weeks Korean ginseng had little effect Lee et al. (2008)
25 (M13F12) Korean ginseng 1.5 gd on warfarin INR and warfarin
metabolism

M male, F female
a
American ginseng root powder contains 26 mg of total ginsenosides. Rb1:Rb2:Rc:Re:Rg1 = 9.6 mg:1 mg:3.05 mg:8.4 mg:1.75 mg
b
Korean ginseng root powder contains 8.93 mg of ginsenoside Rg1
c
Korean red ginseng extract from the 6-year-old root of Korean ginseng contains 100 mg ginsenosides
d
Korean ginseng root powder contains 3 mg and 1.5 mg of Rb1 and Rg1, respectively

13
874 M.-K. Choi, I.-S. Song

Antidiabetic drugs Korean red ginseng, black ginseng, and fermented ginseng
has been proved using in vitro cells systems and diabetic
Anti-diabetic effect of ginseng and its related mechanisms animals (Yuan et al. 2012; Li and Gong 2015). The mecha-
have been investigated to prove the beneficial hypoglyce- nisms underlying the modulation of glucose metabolism by
mic effects of ginseng products and ginsenosides on diabetic ginseng products in diabetic patients would likely be pertur-
animals and patients. The therapeutic potential of several bation of hepatic glucose production and enhancement of
ginseng products from Asian ginseng, American ginseng, glucose uptake in peripheral tissues via glucose transporter

Table 6  Studies investigating the antidiabetic effect of ginseng


Subject characteristics Age Ginseng dose Duration Results References

Ginseng without standard diabetic drugs


DM patients 59.7 ± 7.0 Ginseng 0.2 g/day 8 weeks FBG ↓, HbA1c ↓, OGGT ↓ Sotaniemi et al. (1995)
36 (M16:F20)
DM patients 51.6 ± 10.9 Korean red ginseng 2.7 g/day 24 weeks FBG ↔, HbA1c ↔ Choi et al. (1997)
31 (M15:F16)
DM patients 64.0 ± 7.0 American ginseng 3.0 g/day 8 weeks FBG ↓, FPI ↔, HbA1c ↔ Shishtar et al. (2014)
24 (M13:F11) Insulin resistance ↔
DM patients 51.0 ± 8.5 Panax ginseng powder 2.2 g/ 12 weeks FBG ↓, HbA1c ↔ Ma et al. (2008)
20 (M12:F8) day Insulin resistance ↓
DM patients 64.0 ± 8.7 Korean red ginseng 6.0 g/day 12 weeks FBG ↔, HbA1c ↔, OGGT Vuksan et al. (2008)
19 (M11:F8) ↓, FPI ↓, Insulin resistance ↓
Pre-DM and DM patients 46.0 ± 3.0 Korean red ginseng 3 g/day 4 weeks FBG ↔, FPI ↔ Reeds et al. (2011)
15 (M1:F14) for 2 weeks; 8 g/day for HbA1c ↔
2 weeks
DM patients 56.0 ± 7.2 Fermented Korean red gin- 12 weeks FBG ↓, HbA1c ↓ Kim et al. (2011)
38 (M23:F15) seng 0.78 g/day FPI ↔
DM patients 52.7 ± 11.0 Ginsam 1.5–3.0 g/daya 8 weeks FBG ↓, HbA1c ↓ Yoon et al. (2011)
72 (M44:F28) FPI ↔, Insulin resistance ↓
DM patients 58.8 ± 1.7 Korean red ginseng 5.0 g/day 12 weeks FBG ↔, HbA1c ↔ Bang et al. (2014)
41 (M28:F13) FPI ↔, Insulin resistance ↓
DM patients 53.2 ± 1.8 Fermented Korean red gin- 4 weeks FBG ↓, HbA1c ↔ Oh et al. (2014)
42 (M28:F14) seng 2.7 g/day PPG ↓, AUC​glucose ↓
Impaired fasting glucose 50.5 ± 12.4 Ginseng extract 0.96 g/day 8 weeks Insulin resistance ↓ Park et al. (2014)
11 (M6:F5)
Co-medication of ginseng with diabetic drugs
Pre-DM and DM patients 62.9 ± 12.0 PQS 1.8 g/day + standard 4 weeks FBG ↔, FPI ↔ Zhang et al. (2007),
84 (M51:F33) diabetic ­medicationb Insulin resistance ↔ Shishtar et al. (2014)
No combination effect
DM patients 88.1 ± 17.2 American ginseng extract 12 weeks FBG ↑, HbA1c ↔ Mucalo et al. (2014)
74 (M28:F46) 3.0 g/day + usual diabetic No combination effect
medication (control usual
diabetic medication)
Streptozotocin-induced – Korean red ginseng 2.0 g/ 7 weeks FBG ↓, HbA1c ↔ Nam et al. (2018)
diabetic rats kg/day + metformin 50 mg/ Beneficial effect of
kg/day (control metformin KRG + metformin combina-
50 mg/kg/day) tion
Diabetic db/db mice – Compound K 10 mg/kg + met- 8 weeks Beneficial effect of compound Yoon et al. (2007)
formin 150 mg/kg K + metformin combination
Ginsenoside Re
Pre-DM and DM patients 46.0 ± 3.0 Re 0.2 g/day for 2 weeks; 4 weeks FBG ↔, FPI ↔ Reeds et al. (2011)
15 (M1:F14) 0.5 g/day for 2 weeks HbA1c ↔
DM diabetes mellitus, M male, F female, FBG Fasting blood glucose, FPI Fasting plasma inulin, HbA1c Hemoglobin A1C, OGGT​Oral glucose
tolerance test, PPG Postprandial blood glucose, AUC​glucose Area under plasma concentration of glucose
a
Ginsam: Vinegar extract of Korean ginseng
b
PQS: Extract of American ginseng saponin

13
Interactions of ginseng with therapeutic drugs 875

4 (GLUT4). Recently, PPD-type ginsenosides (Rb1, Rb2, Korean red ginseng exhibiting positive glycemic control
Rc, Rg3, and compound K) and PPT-type ginsenosides was 1.9–3.2% (Awang and Li 2008). Ginsenoside Rb2
(Re, Rg1, and PPT) suppressed hepatic gluconeogenesis is the most effective in decreasing blood glucose levels
via adenosine monophosphate (AMP)-activated protein in streptozotocin-induced diabetic rats (Yokozawa et al.
kinase (AMPK) (Lee et al. 2012; Ryu et al. 2013; Meng 1985); however, Rb2 content in American ginseng root
et al. 2017). Enhancement of GLUT4 expression following and Korean red ginseng was reportedly 0.06% and 0.25%,
treatment with ginseng or ginsenosides in turn increased respectively (Awang and Li 2008).
glucose uptake in adipocytes or skeletal muscle cells (Yuan In two clinical studies involving co-administration of
et al. 2012). ginseng product with other antidiabetic drugs, no addi-
Despite increasing evidence for various therapeu- tional combination effect of ginseng could be found com-
tic effects of ginseng products on diabetic animals, the pared with the standard anti-diabetic drug used as con-
hypoglycemic effect of ginseng product remains non- trol (Table 6). Contrarily, high-dose Korean red ginseng
conclusive (Table 6). A double blind, placebo-controlled and compound K in combination with metformin showed
study of 36 diabetic patients who received 0.2 g/day gin- beneficial effects on FBG in diabetic animals (Table 6).
seng root powder for 8 weeks showed reduction in fast- Reeds et al. (2011) studied the effect of ginsenoside Re
ing blood glucose (FBG) and hemoglobin A1c (HbA1c) in previously diagnosed diabetic patients and prediabetic
levels and an improvement in glucose response to oral subjects but did not report optimistic clinical outcomes
glucose tolerance test (OGGT) (Sotaniemi et al. 1995). (Table 6). As previously mentioned, determining an
However, 31 diabetic patients who underwent long-term optimal dose for ginseng or a single ginsenoside based
treatment for 24 weeks with 2.7 g/day Korean red ginseng on plasma ginsenoside concentration may be useful for
showed no changes in FBG and HbA1c levels (Choi et al. future clinical studies.
1997). Clinical studies, shown in Table 6, varied with
regard to ginseng treatment (i.e., dose, formulation, ori-
gin, and treatment period) and showed partially improved Conclusions and perspectives
clinical outcomes for FBG level and insulin resistance but
was unsuccessful at reducing HbA1c level to the control This comprehensive review on the pharmacokinetic interac-
level. The discrepancy between animal and human stud- tions of ginseng revealed that daily administration of Korean
ies as well as among human studies could be attributed to red ginseng (0.5–3 g extract; dried ginseng, > 60%) did not
different ginsenoside exposures resulting from different cause significant herb–drug interaction with drug-metab-
ginseng treatments. However, plasma ginsenoside con- olizing enzymes (e.g., CYPs) and drug transporters (e.g.,
centrations that exert a hypoglycemic effect have not yet P-gp and OATP1B1). Among different therapeutic drugs co-
been investigated. administered with ginseng, adjuvant chemotherapy, com-
In numerous animal studies showing anti-diabetic prising ginseng (1–3 g extract) and anticancer drugs, facili-
effect of red ginseng extract, red ginseng extract dose tates reduction in CRF and improves the quality of life and
ranges from 200 mg/kg to 2.0 g/kg (i.e., 3–15 mg/kg of emotional scores. It is important to understand the effects of
total ginsenosides) (Xin et al. 2007; Martovetsky et al. ginseng therapy or ginseng combination therapy on diabetic
2013; Nam et al. 2018). In human studies, 2.7–6.0 g/ patients and of ginseng interaction with aspirin or warfarin
day Korean red ginseng extract, usually containing because these effects may vary depending on the composi-
50–100 mg ginsenosides/day, was administered to dia- tion of specific ginsenosides, including Rb1/Rg1 ratio and
betic patients for 4–24 weeks (Table 6). On the basis of Re or Rb2 content, as well as the origin and preparation
the abovementioned ginseng dose and ginsenoside con- method of ginseng products.
tent, the maximum plasma concentration of Rb1 was Although many in vitro cell studies and in vivo animal
36.6 ng/mL after administration of a single oral dose of studies have reported potential ginseng–drug interactions
2 g/kg red ginseng extract to streptozotocin-induced dia- and beneficial effects of ginseng, such results could neither
betic rats (Nam et al. 2018). However, in humans, plasma reflect the actual overall effect of herb–drug interactions in
Rb1 concentration was 2.2 ng/mL after a single dosage of an in vivo system nor extrapolate the therapeutic benefit
Korean red ginseng extract, which increased to 12.2 ng/ of ginseng in clinical settings. Individual case reports of
mL after repeated administration for 2 weeks (Choi et al. herb–drug interaction could not be extrapolated to clinical
2018). After establishing plasma ginsenoside concentra- settings owing to differences in dosage, origins, and formu-
tions, it can be suggested that the anti-diabetic effect of lations of ginseng product in several cases. Therefore, clini-
ginseng is reproducible or comparable with that reported cal results of a standardized ginseng product created using a
in other clinical studies. Another study reported that the pharmacologically effective component and various ginseno-
total ginsenoside content in American ginseng root and sides along with their pharmacokinetic–pharmacodynamic

13
876 M.-K. Choi, I.-S. Song

correlation are of utmost necessity to serve as reproducible Korean volunteers and rats. Evid Based Complement Alternat
and reliable clinical evidence. Med 2016:3908142
Choi S, Oh DS, Jerng UM (2017) A systematic review of the pharma-
cokinetic and pharmacodynamic interactions of herbal medicine
with warfarin. PLoS ONE 12:e0182794
Compliance with ethical standards Choi MK, Jin S, Jeon JH, Kang WY, Seong SJ, Yoon YR, Han YH,
Song IS (2018) Tolerability and pharmacokinetics of ginseno-
Conflict of interest The authors declare no conflict of interest. sides Rb1, Rb2, Rc, Rd, and compound K after single or multiple
administration of red ginseng extract in human beings. J Ginseng
Res. https​://doi.org/10.1016/j.jgr.2018.1010.1006
Council for Responsible Nutrition (2018) Who takes dietary supple-
ments? 2017 CRN consumer survey on dietary supplements. 18
References October 2018. https​://www.crnus​a.org/resou​rces/2017-crn-consu​
mer-surve​y-dieta​rysup​pleme​nts
Agbabiaka TB, Wider B, Watson LK, Goodman C (2017) Concurrent Dong H, Ma J, Li T, Xiao Y, Zhang N, Liu J, Gao Y, Shao J, Jia L
use of prescription drugs and herbal medicinal products in older (2017a) Global deregulation of ginseng products may be a safety
adults: a systematic review. Drugs Aging 34:891–905 hazard to warfarin takers: solid evidence of ginseng–warfarin
Arring NM, Millstine D, Marks LA, Nail LM (2018) Ginseng as a interaction. Sci Rep 7:5813
treatment for fatigue: a systematic review. J Altern Complement Dong WW, Zhao J, Zhong FL, Zhu WJ, Jiang J, Wu S, Yang DC, Li
Med 24:624–633 D, Quan LH (2017b) Biotransformation of Panax ginseng extract
Awang DVC, Li MZC (2008) The pharmacologically active constitu- by rat intestinal microflora: identification and quantification of
ents of white and red ginseng root. Herbalgram 80:38–49 metabolites using liquid chromatography-tandem mass spectrom-
Bang H, Kwak JH, Ahn HY, Shin DY, Lee JH (2014) Korean red gin- etry. J Ginseng Res 41:540–547
seng improves glucose control in subjects with impaired fasting Gui QF, Xu ZR, Xu KY, Yang YM (2016) The efficacy of ginseng-
glucose, impaired glucose tolerance, or newly diagnosed type 2 related therapies in type 2 diabetes mellitus: an updated systematic
diabetes mellitus. J Med Food 17:128–134 review and meta-analysis. Medicine (Baltimore) 95:e2584
Barton DL, Liu H, Dakhil SR, Linquist B, Sloan JA, Nichols CR, Gurley BJ, Gardner SF, Hubbard MA, Williams DK, Gentry WB,
Mcginn TW, Stella PJ, Seeger GR, Sood A, Loprinzi CL (2013) Cui Y, Ang CY (2002) Cytochrome P450 phenotypic ratios for
Wisconsin ginseng (Panax quinquefolius) to improve cancer- predicting herb-drug interactions in humans. Clin Pharmacol
related fatigue: a randomized, double-blind trial, N07C2. J Natl Ther 72:276–287
Cancer Inst 105:1230–1238 Gurley BJ, Gardner SF, Hubbard MA, Williams DK, Gentry WB, Cui
Ben-Eltriki M, Hassona M, Meckling G, Adomat H, Deb S, Tomlinson Y, Ang CY (2005) Clinical assessment of effects of botanical
Guns ES (2019) Pharmacokinetic interaction of calcitriol with supplementation on cytochrome P450 phenotypes in the elderly:
20(S)-protopanaxadiol in mice: determined by LC/MS analysis. St John’s wort, garlic oil, Panax ginseng and Ginkgo biloba.
Eur J Pharm Sci 130:173–180 Drugs Aging 22:525–539
Bilgi N, Bell K, Ananthakrishnan AN, Atallah E (2010) Imatinib and Henderson L, Yue QY, Bergquist C, Gerden B, Arlett P (2002) St
Panax ginseng: a potential interaction resulting in liver toxicity. John’s wort (Hypericum perforatum): drug interactions and
Ann Pharmacother 44:926–928 clinical outcomes. Br J Clin Pharmacol 54:349–356
Bogacz AK, Dziekan KP, Górska-Paukszta M, Kowalska A, Hwang SW, Han HS, Lim KY (2008) Drug interaction between
Mikołajczak P, Ożarowski M, Czerny B (2016) Impact of Panax complementary herbal medicines and gefitinib. J Thorac Oncol
ginseng and ginkgo biloba extracts on expression level of tran- 3:942–943
scriptional factors and xenobiotic-metabolizing cytochrome p450 Jeong JS, Ryu BH, Kim JS, Park JW, Choi WC, Yoon SW (2010)
enzymes. Herba Polonica 62:42–54 Bojungikki-Tang for cancer-related fatigue: a pilot randomized
Chang YD, Smith J, Portman D, Kim R, Oberoi-Jassal R, Rajasekhara clinical trial. Integr Cancer Ther 9:331–338
S, Davis M (2018) Single institute experience with methylpheni- Jeurissen SMF, Buurma-Rethans EJM, Beukers MH, Jansen-Van
date and American ginseng in cancer-related fatigue. Am J Hosp Der Vliet M, Van Rossum CTM, Sprong RC (2018) Consump-
Palliat Care 35:144–150 tion of plant food supplements in the Netherlands. Food Funct
Chen CF, Chiou WF, Zhang JT (2008) Comparison of the pharmaco- 9:179–190
logical effects of Panax ginseng and Panax quinquefolium. Acta Jiang X, Williams KM, Liauw WS, Ammit AJ, Roufogalis BD, Duke
Pharmacol Sin 29:1103–1108 CC, Day RO, Mclachlan AJ (2004) Effect of St. John’s wort and
Chen S, Wang Z, Huang Y, O’barr SA, Wong RA, Yeung S, Chow ginseng on the pharmacokinetics and pharmacodynamics of war-
MS (2014) Ginseng and anticancer drug combination to improve farin in healthy subjects. Br J Clin Pharmacol 57:592–599
cancer chemotherapy: a critical review. Evid Based Complement Jiang R, Dong J, Li X, Du F, Jia W, Xu F, Wang F, Yang J, Niu W,
Alternat Med 2014:168940 Li C (2015) Molecular mechanisms governing different phar-
Choi KT (2008) Botanical characteristics, pharmacological effects and macokinetics of ginsenosides and potential for ginsenoside-per-
medicinal components of Korean Panax ginseng C A Meyer. Acta petrated herb–drug interactions on OATP1B3. Br J Pharmacol
Pharmacol Sin 29:1109–1118 172:1059–1073
Choi KM, Lee EJ, Kim YH, Baik SH, Kim YK, Choi DS (1997) Effects Jiang SL, Liu HJ, Liu ZC, Liu N, Rui L, Kang YR, Ji JG, Zhang C, Hua
of red ginseng on the lipid peroxidation of erythrocyte and anti- BJ, Kang SJ (2017) Adjuvant effects of fermented red ginseng
oxidant superoxide dismutase (SOD) activity in NIDDM patients. extract on advanced non-small cell lung cancer patients treated
J Ginseng Res 21:153–159 with chemotherapy. Chin J Integr Med 23:331–337
Choi ID, Ryu JH, Lee DE, Lee MH, Shim JJ, Ahn YT, Sim JH, Huh Jin S, Jeon JH, Lee S, Kang WY, Seong SJ, Yoon YR, Choi MK, Song
CS, Shim WS, Yim SV, Chung EK, Lee KT (2016) Enhanced IS (2019a) Detection of 13 ginsenosides (Rb1, Rb2, Rc, Rd, Re,
absorption study of ginsenoside Compound K (20-O-beta-(d- Rf, Rg1, Rg3, Rh2, F1, Compound K, 20(S)-Protopanaxadiol, and
Glucopyranosyl)-20(S)-protopanaxadiol) after oral adminis- 20(S)-Protopanaxatriol) in human plasma and application of the
tration of fermented red ginseng extract (HYFRG) in healthy analytical method to human pharmacokinetic studies following 2

13
Interactions of ginseng with therapeutic drugs 877

week-repeated administration of red ginseng extract. Molecules Lee SH, Ahn YM, Ahn SY, Doo HK, Lee BC (2008) Interaction
24:E2618 between warfarin and Panax ginseng in ischemic stroke patients.
Jin S, Lee S, Jeon JH, Kim H, Choi MK, Song IS (2019b) Enhanced J Altern Complement Med 14:715–721
intestinal permeability and plasma concentration of metformin in Lee YH, Lee BK, Choi YJ, Yoon IK, Chang BC, Gwak HS (2009)
rats by the repeated administration of red ginseng extract. Phar- Interaction between warfarin and Korean red ginseng in patients
maceutics 11:E189 with cardiac valve replacement. Int J Cardiol 145:275–276
Jo JJ, Lee S (2017) Investigation of herb-drug interactions between Lee S, Lee MS, Kim CT, Kim IH, Kim Y (2012) Ginsenoside Rg3
Korean red ginseng extract and five Cyp substrates by LC–MS/ reduces lipid accumulation with AMP-activated protein kinase
MS. Mass Spectrom Lett 8:98–104 (AMPK) activation in HepG2 cells. Int J Mol Sci 13:5729–5739
Kim HK (2013) Pharmacokinetics of ginsenoside Rb1 and its metabo- Lee S, Kwon M, Choi MK, Song IS (2018) Effects of red ginseng
lite compound K after oral administration of Korean Red Ginseng extract on the pharmacokinetics and elimination of methotrexate
extract. J Ginseng Res 37:451–456 via Mrp2 regulation. Molecules 23:E2948
Kim DH (2018) Gut microbiota-mediated pharmacokinetics of ginseng Leite PM, Martins MaP, Castilho RO (2016) Review on mechanisms
saponins. J Ginseng Res 42:255–263 and interactions in concomitant use of herbs and warfarin therapy.
Kim HO, Park M, Han J (2011) Effects of fermented Red Ginseng Biomed Pharmacother 83:14–21
supplementation on blood glucose and insulin resistance in type Li KK, Gong XJ (2015) A review on the medicinal potential of Panax
2 diabetic patients. J Korean Soc Food Sci Nutr 40:696 ginseng saponins in diabetes mellitus. RSC Adv 59:47353–47366
Kim H, Nam W, Kim SH, Jang HR, Lee MK, Kim TW, Lee S (2012) Lu JM, Yao Q, Chen C (2009) Ginseng compounds: an update on
Modulatory effects of Korean red ginseng extract (Panax ginseng their molecular mechanisms and medical applications. Curr Vasc
C.A. Meyer) on cytochrome P450 after oral administration to mice Pharmacol 7:293–302
for 14 days. J Life Sci 22:991–998 Ma SW, Benzie IF, Chu TT, Fok BS, Tomlinson B, Critchley LA
Kim D, Zheng YF, Min JS, Park JB, Bae SH, Yoon KD, Chin YW, (2008) Effect of Panax ginseng supplementation on biomarkers
Oh E, Bae SK (2016a) In vitro stereoselective inhibition of gin- of glucose tolerance, antioxidant status and oxidative stress in
senosides toward UDP-glucuronosyltransferase (UGT) isoforms. type 2 diabetic subjects: results of a placebo-controlled human
Toxicol Lett 259:1–10 intervention trial. Diabetes Obes Metab 10:1125–1127
Kim DS, Kim Y, Jeon JY, Kim MG (2016b) Effect of Red Ginseng on Maher JM, Slitt AL, Callaghan TN, Cheng X, Cheung C, Gonzalez FJ,
cytochrome P450 and P-glycoprotein activities in healthy volun- Klaassen CD (2006) Alterations in transporter expression in liver,
teers. J Ginseng Res 40:375–381 kidney, and duodenum after targeted disruption of the transcrip-
Kim MG, Kim Y, Jeon JY, Kim DS (2016c) Effect of fermented red tion factor HNF1alpha. Biochem Pharmacol 72:512–522
ginseng on cytochrome P450 and P-glycoprotein activity in Malati CY, Robertson SM, Hunt JD, Chairez C, Alfaro RM, Kovacs
healthy subjects, as evaluated using the cocktail approach. Br J JA, Penzak SR (2012) Influence of Panax ginseng on cytochrome
Clin Pharmacol 82:1580–1590 P450 (CYP)3A and P-glycoprotein (P-gp) activity in healthy par-
Kim HS, Kim MK, Lee M, Kwon BS, Suh DH, Song YS (2017a) Effect ticipants. J Clin Pharmacol 52:932–939
of red ginseng on genotoxicity and health-related quality of life Martovetsky G, Tee JB, Nigam SK (2013) Hepatocyte nuclear factors
after adjuvant chemotherapy in patients with epithelial ovarian 4 alpha and 1 alpha regulate kidney developmental expression of
cancer: a randomized, double blind, placebo-controlled Trial. drug-metabolizing enzymes and drug transporters. Mol Pharma-
Nutrients 9:E772 col 84:808–823
Kim JH, Yi YS, Kim MY, Cho JY (2017b) Role of ginsenosides, Meng F, Su X, Li W, Zheng Y (2017) Ginsenoside Rb3 strengthens
the main active components of Panax ginseng, in inflammatory the hypoglycemic effect through AMPK for inhibition of hepatic
responses and diseases. J Ginseng Res 41:435–443 gluconeogenesis. Exp Ther Med 13:2551–2557
Kim SJ, Choi S, Kim M, Park C, Kim GL, Lee SO, Kang W, Rhee Mucalo I, Jovanovski E, Vuksan V, Bozikov V, Romic Z, Rahelic D
DK (2018) Effect of Korean Red Ginseng extracts on drug–drug (2014) American ginseng extract (Panax quinquefolius L.) is safe
interactions. J Ginseng Res 42:370–378 in long-term use in type 2 diabetic patients. Evid Based Comple-
Kuo SC, Teng CM, Lee JC, Ko FN, Chen SC, Wu TS (1990) Anti- ment Alternat Med 2014:969168
platelet components in Panax ginseng. Planta Med 56:164–167 Na DH, Ji HY, Park EJ, Kim MS, Liu KH, Lee HS (2011) Evaluation
Kwon HW (2018) Inhibitory Effect of 20(S)-ginsenoside Rg3 on of metabolism-mediated herb–drug interactions. Arch Pharm Res
human platelet aggregation and intracellular Ca(2+) levels via 34:1829–1842
cyclic adenosine monophosphate dependent manner. Prev Nutr Nam SJ, Han YJ, Lee W, Kang B, Choi MK, Han YH, Song IS (2018)
Food Sci 23:317–325 Effect of red ginseng extract on the pharmacokinetics and efficacy
Lau AJ, Woo SO, Koh HL (2003) Analysis of saponins in raw and of metformin in streptozotocin-induced diabetic rats. Pharmaceu-
steamed Panax notoginseng using high-performance liquid tics 10:E80
chromatography with diode array detection. J Chromatogr A Oh MR, Park SH, Kim SY, Back HI, Kim MG, Jeon JY, Ha KC, Na
1011:77–87 WT, Cha YS, Park BH, Park TS, Chae SW (2014) Postprandial
Lau AJ, Toh DF, Chua TK, Pang YK, Woo SO, Koh HL (2009) Anti- glucose-lowering effects of fermented red ginseng in subjects with
platelet and anticoagulant effects of Panax notoginseng: compari- impaired fasting glucose or type 2 diabetes: a randomized, double-
son of raw and steamed Panax notoginseng with Panax ginseng blind, placebo-controlled clinical trial. BMC Complement Altern
and Panax quinquefolium. J Ethnopharmacol 125:380–386 Med 14:237
Laube R, Liu K (2019) An unwanted complement: rare case of poten- Park SH, Oh MR, Choi EK, Kim MG, Ha KC, Lee SK, Kim YG, Park
tial liver injury induced by an interaction between ginseng and BH, Kim DS, Chae SW (2014) An 8-week, randomized, double-
atorvastatin. Br J Clin Pharmacol 85:1612–1613 blind, placebo-controlled clinical trial for the antidiabetic effects
Lee CH, Kim JH (2014) A review on the medicinal potentials of gin- of hydrolyzed ginseng extract. J Ginseng Res 38:239–243
seng and ginsenosides on cardiovascular diseases. J Ginseng Res Park SE, Na CS, Yoo SA, Seo SH, Son HS (2017a) Biotransformation
38:161–166 of major ginsenosides in ginsenoside model culture by lactic acid
bacteria. J Ginseng Res 41:36–42

13
878 M.-K. Choi, I.-S. Song

Park TY, Hong M, Sung H, Kim S, Suk KT (2017b) Effect of Korean Xin HW, Wu XC, Li Q, Yu AR, Zhu M, Shen Y, Su D, Xiong L (2007)
Red Ginseng in chronic liver disease. J Ginseng Res 41:450–455 Effects of Schisandra sphenanthera extract on the pharmacoki-
Qiu F, Zeng J, Liu S, He M, Zhu L, Ye Y, Miao P, Shen S, Jiang J netics of tacrolimus in healthy volunteers. Br J Clin Pharmacol
(2014) Effects of danshen ethanol extract on the pharmacokinetics 64:469–475
of fexofenadine in healthy volunteers. Evid Based Complement Xu T, Jin ZC, Yuan Y, Wei HM, Xu XY, He SL, Chen ST, Hou W,
Alternat Med 2014:473213 Guo QJ, Hua BJ (2016) Ginsenoside Rg3 serves as an adjuvant
Reeds DN, Patterson BW, Okunade A, Holloszy JO, Pololsky KS, chemotherapeutic agent and VEGF inhibitor in the treatment of
Klein S (2011) Ginseng and ginsenoside Re do not improve non-small cell lung cancer: a meta-analysis and systematic review.
β-cell function or insulin sensitivity in overweight and obese Evid Based Complement Alternat Med 2016:7826753
subjects with impaired glucose tolerance or diabetes. Diabetes Yennu S, Tannir NM, Williams JL, Hess KR, Frisbee-Hume S, House
Care 34:1071–1076 HL (2016) Effects of high-dose Asian ginseng (Panax ginseng) to
Ru W, Wang D, Xu Y, He X, Sun YE, Qian L, Zhou X, Qin Y (2015) improve cancer-related fatigue: results of a double-blind, placebo-
Chemical constituents and bioactivities of Panax ginseng (C. A. controlled randomized controlled trial. J Clin Oncol 34:209
Mey.). Drug Discov Ther 9:23–32 Yokozawa T, Kobayashi T, Oura H, Kawashima Y (1985) Studies
Ryu JY, Song IS, Sunwoo YE, Shon JH, Liu KH, Cha IJ, Shin JG on the mechanism of the hypoglycemic activity of ginsenoside-
(2007) Development of the “Inje cocktail” for high-throughput Rb2 in streptozotocin-diabetic rats. Chem Pharm Bull (Tokyo)
evaluation of five human cytochrome P450 isoforms in vivo. Clin 33:869–872
Pharmacol Ther 82:531–540 Yoon SH, Han EJ, Sung JH, Chung SH (2007) Anti-diabetic effects of
Ryu JS, Lee HJ, Bae SH, Kim SY, Park Y, Suh HJ, Jeong YH (2013) compound K versus metformin versus compound K-metformin
The bioavailability of red ginseng extract fermented by Phellinus combination therapy in diabetic db/db mice. Biol Pharm Bull
linteus. J Ginseng Res 37:108–116 30:2196–2200
Seong SJ, Kang WY, Heo JK, Jo J, Choi WG, Liu KH, Lee S, Choi Yoon JW, Kang SM, Vassy JL, Shin HJ, Lee YH, Ahn HY, Choi SH,
MK, Han YH, Lee HS, Ohk B, Lee HW, Song IS, Yoon YR Park KS, Jang HC, Lim S (2011) Efficacy and safety of ginsam,
(2018) A comprehensive in vivo and in vitro assessment of the a vinegar extract from Panax ginseng, in type 2 diabetic patients:
drug interaction potential of red ginseng. Clin Ther 40:1322–1337 results of a double-blind, placebo-controlled study. J Diabetes
Shih DQ, Bussen M, Sehayek E, Ananthanarayanan M, Shneider BL, Investig 3:309–317
Suchy FJ, Shefer S, Bollileni JS, Gonzalez FJ, Breslow JL, Stoffel Yuan CS, Wei G, Dey L, Karrison T, Nahlik L, Maleckar S, Kasza K,
M (2001) Hepatocyte nuclear factor-1alpha is an essential regu- Ang-Lee M, Moss J (2004) Brief communication: American gin-
lator of bile acid and plasma cholesterol metabolism. Nat Genet seng reduces warfarin’s effect in healthy patients: a randomized,
27:375–382 controlled Trial. Ann Intern Med 141:23–27
Shishtar E, Sievenpiper JL, Djedovic V, Cozma AI, Ha V, Jayalath Yuan HD, Kim JT, Kim SH, Chung SH (2012) Ginseng and diabetes:
VH, Jenkins DJ, Meija SB, De Souza RJ, Jovanovski E, Vuksan the evidences from in vitro, animal and human studies. J Ginseng
V (2014) The effect of ginseng (the genus panax) on glycemic Res 36:27–39
control: a systematic review and meta-analysis of randomized Yun TK, Choi SY, Yun HY (2001) Epidemiological study on cancer
controlled clinical trials. PLoS ONE 9:e107391 prevention by ginseng: are all kinds of cancers preventable by
Sotaniemi EA, Haapakoski E, Rautio A (1995) Ginseng therapy in non- ginseng? J Korean Med Sci 16(Suppl):S19–S27
insulin-dependent diabetic patients. Diabetes Care 18:1373–1375 Zhang Y, Lu S, Liu YY (2007) Effect of panax quinquefolius saponin
Teng CM, Kuo SC, Ko FN, Lee JC, Lee LG, Chen SC, Huang TF on insulin sensitivity in patients of coronary heart disease with
(1989) Antiplatelet actions of panaxynol and ginsenosides isolated blood glucose abnormality. Zhongguo Zhong Xi Yi Jie He Za Zhi
from ginseng. Biochim Biophys Acta 990:315–320 27:1066–1069
Tian Z, Pang H, Du S, Lu Y, Zhang L, Wu H, Guo S, Wang M, Zhang Zhang R, Jie J, Zhou Y, Cao Z, Li W (2009) Long-term effects of
Q (2017) Effect of Panax notoginseng saponins on the pharma- Panax ginseng on disposition of fexofenadine in rats in vivo. Am
cokinetics of aspirin in rats. J Chromatogr B Analyt Technol J Chin Med 37:657–667
Biomed Life Sci 1040:136–143 Zhou J, Wu J, Wu CY, Long F, Shen H, Zhang W, Li SL (2019a) Herb–
Tian Z, Pang H, Zhang Q, Du S, Lu Y, Zhang L, Bai J, Li P, Li D, Zhao drug interaction: a case study of effects and involved mechanisms
M, Chen X (2018) Effect of aspirin on the pharmacokinetics and of cisplatin on the pharmacokinetics of ginsenoside Rb1 in tumor-
absorption of panax notoginseng saponins. J Chromatogr B Analyt bearing mice. Biomed Pharmacother 110:95–104
Technol Biomed Life Sci 1074–1075:25–33 Zhou L, Chen L, Wang Y, Huang J, Yang G, Tan Z, Wang Y, Liao J,
Vuksan V, Sung MK, Sievenpiper JL, Stavro PM, Jenkins AL, Di Zhou G, Hu K, Li Z, Ouyang D (2019b) Impact of NR1I2, adeno-
Buono M, Lee KS, Leiter LA, Nam KY, Arnason JT, Choi M, sine triphosphate-binding cassette transporters genetic polymor-
Naeem A (2008) Korean red ginseng (Panax ginseng) improves phisms on the pharmacokinetics of ginsenoside compound K in
glucose and insulin regulation in well-controlled, type 2 diabetes: healthy Chinese volunteers. J Ginseng Res 43:460–474
results of a randomized, double-blind, placebo-controlled study of Ziemann J, Lendeckel A, Muller S, Horneber M, Ritter CA (2019)
efficacy and safety. Nutr Metab Cardiovasc Dis 18:46–56 Herb–drug interactions: a novel algorithm-assisted information
Won HJ, Kim HI, Park T, Kim HK, Jeon H, Ha SJ, Hyun JM, Jeong system for pharmacokinetic drug interactions with herbal supple-
A, Kim JS, Park YJ, Eo YH, Lee J (2019) Non-clinical pharma- ments in cancer treatment. Eur J Clin Pharmacol 75:1237–1248
cokinetic behavior of ginsenosides. J Ginseng Res 43:354–360
Wu X, Ma J, Ye Y, Lin G (2016) Transporter modulation by Chinese Publisher’s Note Springer Nature remains neutral with regard to
herbal medicines and its mediated pharmacokinetic herb–drug jurisdictional claims in published maps and institutional affiliations.
interactions. J Chromatogr B Analyt Technol Biomed Life Sci
1026:236–253

13

You might also like