Startseite An open-label, single dose, safety and pharmacokinetic study of Withania somnifera root extract in healthy volunteers
Artikel Open Access

An open-label, single dose, safety and pharmacokinetic study of Withania somnifera root extract in healthy volunteers

  • Eshita Sharma , Gayatri Ganu , Ketan Kshirsagar , Ashwin Shah , Umakant Mahale , Anirudh Mehta und Sujit Nair ORCID logo EMAIL logo
Veröffentlicht/Copyright: 19. Februar 2025
Veröffentlichen auch Sie bei De Gruyter Brill

Abstract

Objectives

Withania somnifera (WS), also known as Ashwagandha, is a health-beneficial Ayurvedic medicinal plant with great potential as an adaptogen with rejuvenating and anti-aging effects. However, studies investigating pharmacokinetics (PK), safety, and tolerability of WS on humans are limited. The present study evaluated PK, safety, and tolerability of WS root extract (2.5 % total withanolides) capsules upon oral administration of two capsules of 200 mg each (total 400 mg) in healthy male and female volunteers.

Methods

An open label, single dose, clinical design comprising healthy volunteers was employed. The study evaluated PK parameters of the four bioactive constituents viz. withanoside IV, withaferin A, 12-deoxy-withastramonolide, and withanolide A in WS root extract after analysis of plasma using a validated UHPLC-MS/MS method. Further, safety and tolerability assessment for vital signs, testing for organ function, urine examination, X-ray, ECG, as well as adverse events profile were also investigated.

Results

After oral administration of 2 WS capsules (200 mg each), the participants reported normal physical, hematological, and biochemical parameters with no abnormalities in safety metrics. For the four bioactives, the exposure parameters range between 0.472 and 4.468 ng/mL (Cmax), 1.000–1.416 h (Tmax), and 2.051–13.319 ng/mL*h (AUC 0-t). Further, t1/2 (1.696–4.377 h), lambda_z (0.141–0.282 L/h), Cl/F (0.065–0.954 mg/(ng/ml)/h), AUMC 0-inf_obs (21.720–80.485 ng/mL*hˆ2) and MRT 0-inf_obs (3.680–7.516 h) also differed for each bioactive.

Conclusions

The present study elucidated the PK of WS and showed that healthy male and female volunteers may safely consume WS capsules at a dose of 400 mg (2 capsules of 200 mg) without any harmful effects.

Introduction

Ashwagandha or Withania somnifera L. Dunal (WS) due to its numerous health benefits has been used since ancient times, and is generally located in South Asia, Central Asia, and Africa arid regions [1]. WS has been included as an important medicinal plant in WHO monographs [2], is the third highest selling product in US market after cannabidiol and elderberry [3] and declared as a primary concern for mechanistic investigation in humans by The National Center for Complementary and Alternative Medicines (NCCAM) of the US-NIH (National Institutes of Health) [4]. WS has been acknowledged for its numerous health benefits such as immunomodulation, anti-inflammatory, anti-aging, neuroprotective, antioxidant, anticancer, anti-stress, anti-anxiety, and anti-hypertensive properties [5], [6], [7], [8], [9], [10], [11], [12], [13].

WS possesses bioactive compounds like steroidal lactones (withanolides), alkaloids, sitoindosides, flavonoids, and tannins [1]. The bioactive phytoconstituents are mainly present in the roots of WS plant, however other parts also possess bioactive compounds [12]. The bioactive constituents present in roots include withaferin A, withanolides A and B, 12-deoxy withastromonolide, and withanosides. The withanolides are responsible for various pharmacological activities in different concentrations varying from micrograms to milligrams [6]. Withaferin A possesses anticancer, anti-inflammatory, anti-parasitic, and hepatoprotective potential [14], 15]. Withanolide A has attracted attention as a potential candidate for the therapeutic treatment of neurodegenerative disorders including Alzheimer’s disease, Parkinson’s disease, and cognitive function impairment. The molecular mechanism studies on withaferin A and withanolides revealed the modulation of targets viz. enzymes, transcriptional factors, cytokines, growth factors and other targets suggesting promising candidates in cancer and neurological disorders [16], [17], [18].

Pharmacokinetic (PK) studies on biochemical components provide valuable insights into their metabolism, dosage, as well as guidance for their clinical use. We have previously discussed the role of clinical pharmacometrics in cancer precision medicine [19], 20] We have previously reported the clinical pharmacokinetics of dietary polyphenols [21], and the nutraceutical sulforaphane [22], the monoclonal antibody olaratumab [23] and the immune checkpoint inhibitor nivolumab in cancer translational medicine [24]. Despite various mechanistic studies, there are very few reports on human PK of WS extracts containing withanolides and withanosides [3], 19], 25]. However, the absorption of the bioactive compounds, metabolism, tissue distribution, and their excretion from the body when administered orally is of great relevance [19], 26]. Kim et al. [19] reported that the concentration of withanosides in WS extract was crucial for ensuring oral bioavailability. The higher the extract strength, the longer was the half-life, thus, higher was the mean residence time [19]. Alluri et al. [25] demonstrated increased relative absorption, effective relative bioavailability, and longer elimination half-life of sustained-release WS root extract capsules.

Although several pre-clinical and clinical studies have documented the biological and therapeutic properties of WS bioactive compounds, however their PK, safety, and tolerability profiles have been less explored. Nevertheless, the safety profile of WS has been a major concern worldwide for the nutraceutical and functional food industry. Previously published reports on safety assessment revealed a safe escalating dose of WS aqueous extract from 750-1,250 mg/day [27] and root extract at 600 mg/day [28] and 1,000 mg/day in male and female participants [29], 30]. On the contrary, WS extract (600 mg daily, 50 participants, 8 weeks) when tested for anxiety reported side effects like drowsiness, lesser appetite, minor dizziness, heavy head, unclear vision in memory, mild transient fever, asthenia, and cough in subclinical hypothyroid patients [31]. In a randomized double-blind, placebo controlled study of safety and efficacy on 60 participants administered Ashwagandha capsules containing 300 mg high-concentration extract of WS roots for 60 days, there were mild side-effects along with no adverse effects [32].

Variation in concentration of withanolides in different products containing WS extract has been noticed globally. Hence, in-depth studies are needed to understand the clinical safety of optimized WSE formulation in human populations. Thus, the objective of the current study was to investigate the oral pharmacokinetics of active withanolides and withanosides in human plasma after administration of standardized WS root extract capsules in healthy volunteers. In this study, an open label, single dose design was employed to evaluate the PK profiles of withanolides and withanosides in human plasma using a validated bioanalytical method. Further, safety and tolerability of WS extract capsules in human subjects were also assessed during the duration of the PK study.

Materials and methods

Chemicals and reagents

Withanoside IV (purity≥95.00 %) and withanolide A (purity≥98.00 %) were purchased from USP (Rockville, MD, USA). Withaferin-A (purity≥94.80 %) and 12-deoxy-withastramonolide (purity≥96.10 %) were procured from Chromadex (Los Angeles, CA, USA). Fluoxymesterone (FMC, internal standard, IS) with a purity≥98.82 % was acquired from Clearsynth, Mumbai, Maharashtra, India. MS grade water, methanol, and acetonitrile were purchased from JT Baker, Radnor, PA, USA. Ammonium formate was procured from VWR Chemicals, OH, USA. LongeFera™ capsules (each containing 200 mg of 2.5 % W. somnifera root extract) is marketed globally by Phytoveda, USA and Phytoveda Pvt. Ltd., India.

Study design

The present study was performed at Lokmanya Medical Research Centre, Chinchwad, Pune, Maharashtra, India, which was an open label, single-dose study wherein each participant was given a dose of two capsules of WS extract each containing 2.5 % w/w total withanolides i.e. 5 mg/capsule total withanolides and plasma samples were withdrawn for PK analyses upto 24 h. A total of 13 participants were screened with age between 18 and 45 years and 12 participants (6 male + 6 female) were enrolled in the study (Figure 1). Each selected participant signed the informed consent form indicating their voluntary consent to take part in the study.

Figure 1: 
CONSORT study flow diagram of pharmacokinetics and clinical safety of WS root extract in healthy volunteers.
Figure 1:

CONSORT study flow diagram of pharmacokinetics and clinical safety of WS root extract in healthy volunteers.

Ethical approval

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Ethical Committee of Lokmanya Medical Research Centre, Chinchwad, Pune, Maharashtra, India (ECR/175/Inst/MH/2013/RR-19). The research was conducted in compliance with the Indian Council of Medical Research’s (ICMR) and biomedical research ethical guidelines for human participants. After being approved from the Ethics Committee, the registration of the trial was done in Clinical Trial Registry of India (CTRI/2023/09/058168).

Participant screening

The screening of participants was done within 21 days prior to dosing once informed consent was received. The informed consent form was used in the language best understood by the subjects (Marathi/Hindi). During this process, participants were given information regarding the objective and design of the study, possible risks, and benefits of participation in the study. Participants were free to ask questions to fully understand the study and discuss any concerns. The willing participants indicated their compliance for participation through reading, signing, and dating the consent document. A signed document copy was handed over to the participant and a duplicate copy was placed securely in the Informed Consent Form file.

Selection of study participants

Criteria, i.e., inclusion and exclusion were used to determine the eligibility of the participants for the selected study as follows:

Inclusion criteria

Healthy individuals, i.e., 18–45 years old were included. Females with a negative pregnancy test or those with childbearing potential who were willing to take appropriate contraceptive measures during the study and for 7 days after the study were included. Male participants who agreed to take proper contraceptive precautions and abstained from giving of sperm during the study and for 7 days after the study completion were included. Participants with BMI between 18.5 and 30.0 kg/m2 with a body mass not less than 50.00 kg were included. Participants needed to be in regular health, which was established on personal medical history and clinical examination test results. A significant electrocardiogram, chest X-ray, negative result of alcohol breath test was considered for the study. Other necessary conditions include active communication, written notified consent, availability during the entire study and ability to fast for at least 14 h as well as consumption of standard meals.

Exclusion criteria

Participants with hypersensitivity to WS or other related drugs, incapable of understanding the consent information, having a history or presence of any serious disorder, or on any treatment which could bring about initiation or suppression of the hepatic microsomal enzyme system were excluded. Participants noting the presence of drug abuse, asthma, or any allergic responses, gastric or duodenal ulcers, easy bruising, bleeding, thyroid, adrenal dysfunction, organic intracranial lesion, cancer, intake of anticonvulsants, or barbiturates for one month beforehand or throughout the study, smokers, and those with major illness during the 90 days before screening, with positive HIV, Hepatitis B, and C were omitted from the study. Pregnant, nursing women with postmenopausal spontaneous amenorrhea, bilateral oophorectomy with or without a hysterectomy with absence of bleeding for 3 or 6 months were excluded from the study.

Study procedure

Participants were admitted to the study center on day −1 (housing) which was before the day of dose administration. Each participant undertook a complete general clinical and vital sign check. On day 01, vital signs were assessed. An alcohol breath test was performed. Before dosing, participants fasted for at least 10 h until 4 h following dosing. After dosing, about 200 mL of ambient temperature water was given at about every hour starting from 01-h post dose till the end. Water was restricted from at least 01 h before dosing until at least 01-h post-dose (no fluid, except water given with dosing). Free access to water was allowed outside this interval.

Collection and handling of plasma samples

After the overnight fasting for 10 h, participants were given a single dose of 400 mg (two 200 mg capsules in sequence) of WS root extract capsule orally in a sitting posture with 240 ± 2 mL of water at room temperature. Participants swallowed the capsules without being chewed, crushed, or bitten in the presence of the principal investigator. The time of dosing on Day 01 was recorded as the reference time for all subsequent PK blood sample collections. The collection of PK blood samples was performed at pre-dose (within 45 min before oral administration) and post-dose at time points i.e., 0.25 (15 min), 0.50 (30 min), 0.75 (45 min), 1.00, 1.50, 2.00, 3.00, 4.00, 6.00, 9.00, 12.00, and 24.00 h, for an overall of 13 samples per participant (Figure 2). PK blood samples were collected using a venous cannula inserted in a forearm vein or dorsal aspect of the hand. An intravenous indwelling cannula was used in situ if required by injecting 0.3 mL of normal saline or heparinized saline (to prevent the cannula from clogging). At every sampling, 3 mL of the blood was collected in a pre-labeled K2-EDTA collection tube as an anticoagulant. 10 mL blood was withdrawn for safety assessment at screening and 10 mL at the end of the study. The centrifugation of blood samples was performed at 4,000 rpm at 4 °C ± 2 °C for 10 min within 30 min after collection. Further, the plasma was transferred into labelled polypropylene tubes in duplicates with each tube highlighting all details and stored in a freezer at −80 °C ± 10 °C at the analytical facility until analysis. The analyses of plasma samples were performed for WS bioactives via an ultra-high pressure liquid chromatography-mass spectrometry/mass spectrometry (UHPLC-MS/MS) method.

Figure 2: 
Schematic diagram representing procedure of clinical pharmacokinetics.
Figure 2:

Schematic diagram representing procedure of clinical pharmacokinetics.

Pharmacokinetic analyses

Pharmacokinetic parameters like maximum observed concentration (C max ), maximum observed time (T max ), area under the curve (AUC 0-t ), (AUC 0-∞ ), half-life (t 1/2 ), and elimination rate constant (K el ) were calculated from the plasma concentration vs. time data. Estimation of C max as well as T max was directly done from the experimental results. K el was estimated from the linear regression slope of log concentrations as a time function. The t1/2 was calculated as 0.693/K el . The calculation of AUC(0–t) and AUC(0–∞) was performed from the linear curve, and by use of Ct/λ, where Ct was the final measurable concentration, respectively. Oral clearance (Cl/F) was acquired from Cl/F=Dose/AUC(0–∞). The mean residence time (MRT) was estimated as AUMC(0–∞)/AUC(0–∞). Each sample underwent triplicate analyses, with mean ± standard deviation (SD) representing the findings. The preparation of mean plasma concentration vs. time curve was completed employing GraphPad Prism 8.2.

Bioanalytical analysis

WS root extract capsules were quantified for total withanolides and withanosides by following the method of Vaidya et al. [29] employing HPLC with some modifications. Further, the preparation and analyses of plasma samples were performed using the UHPLC-MS/MS method of Vaidya et al. [33] with minor adjustments. A Shimadzu UHPLC system including degasser DGU-20A5R, LC-30AD pump, autosampler SIL-30AC, and CTO-20AC column oven was used. The column used was Agilent C18 ZORBAX Eclipse Plus (4.6 × 100 mm, 3.5 μm) fitted with a Phenomenex guard column. In UHPLC-MS/MS, the analyses of plasma samples were done in positive ion mode and ESI contact temperature was set at 300 °C. The nebulizing gas, heating gas and drying gas flow rate was 3.00 L/min, 10.00 L/min, and 10.00 L/min, respectively. All the analyses were interpreted using Lab Solutions software version 5.95.

Safety assessment

Safety assessment was performed at screening and at the end of the study. Vital signs assessment, clinical examination, clinical laboratory test, urine analysis, ECG, monitoring, and capturing of adverse events/serious adverse events were performed. Additionally, tolerability was also assessed. Blood volumes withdrawn for this assessment have been mentioned in Section Study procedure.

Statistical analysis

Modeling of noncompartmental pharmacokinetics was performed through software PK solver 2.0, USA, to illustrate the concentration-time profiles of WS extract (2.5 %) 400 mg capsules for each healthy subject. PK parameters were representative of the averages of six healthy male and female volunteers, calculated separately, along with their respective standard deviations. For safety analysis, normality of the study data was calculated by Kolmogorov-Smirnov test. Data for clinical laboratory testing was analyzed by Student’s t-test and vital signs by one-way ANOVA. Data for ECGs and chest X-rays were analyzed by Chi-square test. All the statistical analyses were performed using SPSS version 10.00.

Results

Demographic characteristics

Thirteen volunteers were screened for the study; one was excluded from the study for not complying with the inclusion criteria; hence, 12 volunteers were included in the study. All the 12 participants (6 males and 6 females) completed the study and data was evaluated. The CONSORT study flowchart has been depicted in Figure 1. The average age of the participants recorded was 27.58 years. Male participants had a mean weight, height and BMI of 62.33 kg, 165.55 cm and 22.67 kg/m2, respectively. Female participants recorded a mean weight, height and BMI of 64.23 kg, 157.83 cm and 25.70 kg/m2, respectively (Supplemental Table S1). All the values were within clinically acceptable ranges. All the participants involved in the study were non-drinkers and non-smokers. The data was examined via the Student’s t-test at p<0.05.

Quantification of total withanolides in WS capsules

Capsules contained about 200 mg of withanolide extract containing 2.5 % of total withanolides and this accounts for 5 mg/capsule of total withanolides. The total withanolides content was quantified in WS capsules employing HPLC-PDA method and results revealed that total content of withanolides was not less than 5 mg/capsule (Table 1, Figure 3). The content of withaferin A was 1.52 mg/capsule, whereas withanoside IV, withanolide A and 12-deoxy-withastramonolide content calculated was 1.21, 0.71 and 0.41 mg/capsule, respectively. The contents of withaferin A, withanoside IV, withanolide A and 12-deoxy-withastramonolide administered in this study were 1.52, 1.21, 0.71, 0.41 mg, respectively.

Table 1:

HPLC-quantification of total withanolides in WS root extract capsules.

Active compounds Batch no: HCZ012 (n=10)
Content (mg/capsule)
Withanoside IV 1.21 ± 0.04a
Withanoside V 1.31 ± 0.01
Withaferin A 1.52 ± 0.01
12-Deoxy-withastramonolide 0.41 ± 0.01
Withanolide A 0.71 ± 0.01
Withanone 0.02 ± 0.01
Withanolide B 0.20 ± 0.01
mg/capsule 5.38 ± 0.10
  1. aAll the readings were taken in triplicate (mean ± standard deviation).

Figure 3: 
HPLC chromatogram of bioactive constituents present in root extract of WS. (A) Standard mix; (B) root extract sample.
Figure 3:

HPLC chromatogram of bioactive constituents present in root extract of WS. (A) Standard mix; (B) root extract sample.

Assessment of in vivo pharmacokinetic parameters

Data from all the participants was included in the PK analyses. The active constituents were quantified simultaneously using a validated UHPLC-MS/MS bioanalytical method after single dose administration of WS root extract in male and female subjects. The four constituents were quantified at the first-time point i.e., 15 min. LC-MS/MS chromatograms of four bioactive compounds upon oral administration of WS root extract capsules at a dose of 400 mg in male and female participants is shown in Supplemental Figure S1. The mean plasma concentration vs. time curves of four constituents in male and female subjects are given in Figure 4.

Figure 4: 
Average plasma concentration vs. time curves of four constituents (A) overlay, average plasma concentration vs. time curves of four constituents quantified in male volunteers; (B–E) individual constituents after oral administration of WS extract (not less than 2.5 %) capsule at the dose of 400 mg in male volunteers (mean ± SD, n=06 male participants); (F) overlay, average plasma concentration vs. time curves of four constituents quantified in female volunteers; (G–J) individual constituents after oral administration of WS extract (not less than 2.5 %) capsule at the dose of 400 mg in female volunteers (mean ± SD, n=06 female participants).
Figure 4:

Average plasma concentration vs. time curves of four constituents (A) overlay, average plasma concentration vs. time curves of four constituents quantified in male volunteers; (B–E) individual constituents after oral administration of WS extract (not less than 2.5 %) capsule at the dose of 400 mg in male volunteers (mean ± SD, n=06 male participants); (F) overlay, average plasma concentration vs. time curves of four constituents quantified in female volunteers; (G–J) individual constituents after oral administration of WS extract (not less than 2.5 %) capsule at the dose of 400 mg in female volunteers (mean ± SD, n=06 female participants).

In males, C max for withanoside IV was 0.472 ng/mL, withanolide A was 2.981 ng/mL, withaferin A was 3.561 ng/mL and 12-deoxy withastramonolide was 4.468 ng/mL, and the observed T max was 1.333 h for withanoside IV, 1.416 h for withanolide A, 1.000 h for withaferin A and 1.083 h for 12-deoxy withastramonolide (Table 2). Further, AUC(0–24 h) was observed as 2.051 ng/mL*h for withanoside IV, 8.085 ng/mL*h for withaferin A, 13.319 ng/mL*h for 12-Deoxy-withastramonolide and 10.958 ng/mL*h for withanolide A. The (AUC0-t/AUC0-∞) ratio was 0.715 for withanoside IV, 0.838 for withanolide A, 0.802 for withaferin A, and 0.919 for 12-deoxy-withastramonolide. Further, t 1/2 was shortest for 12-deoxy withastramonolide along with shortest oral clearance rate (Table 2). However, in case of female subjects, Table 3 shows that the Cmax and Tmax was found to be 0.673 ng/mL and 1.333 h for withanoside IV, 1.407 ng/mL and 1.166 h for withanolide A, 1.927 ng/mL and 0.916 h for withaferin A and 3.214 ng/mL and 1.071 h for 12-deoxy withastramonolide. AUC(0–24h) and AUC0-t/AUC0-∞ ratio was reported to be the highest for 12-deoxy withastramonolide and lowest for withanolide A (Tables 2 and 3). The LLOQ value of all the constituents was found to be less than C max /10 ratio in both male and female participants indicating that the developed method was sensitive for the quantification of constituents in the human plasma samples. Moreover, in female subjects, t 1/2 of withanoside IV (4.377 h), was found to be greater than that of withanolide A (3.782 h), followed by withaferin A (1.696 h), and 12-deoxy withastramonolide (2.086 h) with oral clearance of 0.616 mg/(ng/mL)/h for withanoside IV, 0.340 mg/(ng/mL)/h for withanolide A, 0.818 mg/(ng/mL)/h for withaferin A, and 0.116 mg/(ng/mL)/h for 12-deoxy withastramonolide (Table 3). In male subjects, withanolide A was detected in plasma for a longer time up to 15 h, whereas withanoside IV, withaferin A and 12-deoxy withastramonolide were detected in plasma up to 12 h. Further in female participants, withanoside IV and 12-deoxy withastramonolide were present in plasma up to 9 h; withaferin A and withanolide A were detected up to 6 h which were lesser plasma time compared to male subjects. The longer duration in plasma indicates lower elimination rate of bioactive constituents attributed to biotransformation, or tissue binding after passing through intestinal wall and liver. Further, hydroxylation, hydrogenation and hydrolysis of compounds in the intestine and liver could be a reason for their extended duration in the body.

Table 2:

Pharmacokinetic analysis of withanolides and withanosides in healthy male participants upon single oral administration of WS (not less than 2.5 %) extract at a dose of 400 mg.

PK Parameters Unit WSIV WLA WFA WSL
Lambda_z 1/h 0.141 ± 0.025 0.155 ± 0.031 0.177 ± 0.047 0.282 ± 0.028
t 1/2 h 5.030 ± 0.997 4.585 ± 0.793 4.108 ± 0.910 2.470 ± 0.241
T max h 1.333 ± 0.577 1.416 ± 0.645 1.000 ± 0.524 1.083 ± 0.465
C max ng/mL 0.472 ± 0.112 2.981 ± 0.895 3.561 ± 1.624 4.468 ± 0.918
Tlag H 0.000 ± 0.000 0.000 ± 0.000 0.000 ± 0.000 0.000 ± 0.000
Clast_obs/C max 0.243 ± 0.036 0.101 ± 0.010 0.0968 ± 0.030 0.076 ± 0.021
AUC0-t ng/mLah 2.051 ± 0.143 10.958 ± 4.800 8.085 ± 3.871 13.319 ± 2.585
AUC 0-inf_obs ng/mLah 2.877 ± 0.160 12.894 ± 5.036 9.968 ± 4.557 14.494 ± 2.857
AUC 0-t/0-inf_obs 0.715 ± 0.070 0.838 ± 0.042 0.802 ± 0.037 0.919 ± 0.010
AUMC 0-inf_obs ng/mLahˆ2 21.720 ± 4.519 80.485 ± 24.671 50.490 ± 24.220 53.474 ± 12.056
MRT 0-inf_obs h 7.516 ± 1.218 6.405 ± 0.690 5.019 ± 0.760 3.680 ± 0.292
Vz/F_obs (mg)/(ng/mL) 6.884 ± 1.050 0.936 ± 0.370 2.429 ± 0.965 0.232 ± 0.045
Cl/F_obs (mg)/(ng/mL)/h 0.954 ± 0.054 0.136 ± 0.040 0.434 ± 0.222 0.065 ± 0.011
  1. aData represented as mean ± SD, (n=06 male subjects). WSIV, Withanoside IV; WLA, Withanolide A; WFA, Withaferin A; WSL, 12-deoxy withastramonolide; C max  , maximal observed concentration; T max , maximum observed time; t 1/2 , half-life; K el , rate of elimination; AUC, area under the curve; AUMC, area under the moment curve; MRT, mean residence time; Cl/F, mean apparent clearance; Vz/F, mean apparent volume of distribution.

Table 3:

Pharmacokinetic analysis of withanolides and withanosides in healthy female participants upon single oral administration of WS (not less than 2.5 %) extract at a dose of 400 mg.

PK Parameters Unit WSIV WLA WFA WSL
Lambda_z 1/h 0.160 ± 0.024a 0.194 ± 0.041 0.423 ± 0.084 0.334 ± 0.031
t 1/2 h 4.377 ± 0.622 3.782 ± 1.089 1.696 ± 0.375 2.086 ± 0.192
T max h 1.333 ± 0.577 1.166 ± 0.408 0.916 ± 0.129 1.071 ± 0.491
C max ng/mL 0.673 ± 0.223 1.407 ± 0.525 1.927 ± 0.484 3.214 ± 0.640
Tlag H 0.000 ± 0.000 0.000 ± 0.000 0.000 ± 0.000 0.000 ± 0.000
Clast_obs/C max 0.493 ± 0.134 0.387 ± 0.126 0.220 ± 0.044 0.140 ± 0.058
AUC0-t ng/mLah 2.540 ± 0.485 2.761 ± 1.116 3.567 ± 1.072 7.618 ± 2.874
AUC 0-inf_obs ng/mLah 4.513 ± 0.700 5.707 ± 2.702 4.555 ± 1.125 8.988 ± 3.499
AUC 0-t/0-inf_obs 0.561 ± 0.058 0.500 ± 0.057 0.775 ± 0.057 0.848 ± 0.026
AUMC 0-inf_obs ng/mLahˆ2 32.018 ± 6.558 34.797 ± 22.903 12.405 ± 2.656 30.661 ± 13.059
MRT 0-inf_obs h 7.105 ± 0.998 5.789 ± 1.220 2.770 ± 0.480 3.395 ± 0.232
Vz/F_obs (mg)/(ng/mL) 3.894 ± 0.835 1.757 ± 0.664 2.038 ± 0.749 0.351 ± 0.171
Cl/F_obs (mg)/(ng/mL)/h 0.616 ± 0.095 0.340 ± 0.163 0.818 ± 0.206 0.116 ± 0.052
  1. aData represented as mean ± SD, (n=06 female subjects); WSIV, Withanoside IV; WLA, Withanolide A; WFA, Withaferin A; WSL, 12-Deoxy withastramonolide; C max  , maximal observed concentration; T max , maximum observed time; t 1/2 , half-life; K el , rate of elimination; AUC, area under the curve; AUMC, area under the moment curve; MRT, mean residence time; Cl/F, mean apparent clearance; Vz/F, mean apparent volume of distribution.

Hematological and biochemical analyses

Hematological and biochemical investigations were performed at the time of screening and study completion (Supplemental Tables S2 and S3). The total protein and albumin levels significantly decreased but remained within normal physiological ranges (Supplemental Table S4). Potassium and urea levels increased however, they were found to be within the normal range and were not clinically significant. No statistically or clinically significant changes were detected in other hematological and biochemical parameters, all of which remained within typical constraints both prior to and following the use of the interventional product (Supplemental Tables S2 and S3).

Assessment of urine analysis

Physicochemical characteristics and microscopic examination as well as color, pH, transparency, specific gravity, blood in urine, protein, glucose, ketone bodies, bilirubin, urobilinogen, RBC, WBC, pus cells, etc. were evaluated and observed within normal confines at the screening and end of the experiment (Supplemental Table S5). No adverse effects were observed after the oral administration of WS root extract capsules.

Vital signs assessment

Changes in vital signs were assessed on the 12 healthy volunteers during screening, housing and after the oral administration of WS root extract capsules (Supplemental Table S6). Results demonstrated that at baseline, blood pressure, pulse rate, respiratory rate, and oral temperature all were within the expected normal range. No clinical as well as statistically significant changes were observed in blood pressure, pulse rate, respiratory rate, and oral body temperature after treatment, hence no adverse effects were reported.

Assessment of chest X-ray and ECG

In chest X-ray and ECG examinations, no clinical or statistically significant abnormalities or changes were detected in QT interval at p˂0.05 at screening and at study completion. Results demonstrated that administered compounds of WS root extract capsules did not affect QT intervals.

Assessment of adverse events and tolerability

All 12 subjects showed excellent tolerability to the investigational capsules.

Discussion

WS or Ashwagandha or Indian cherry has been well recognized for its health attributes since ancient times [34]. WS in Ayurveda, the traditional Indian system of medicine, is recognized as “Avarada” indicating regeneration or youthfulness and as ‘Rasayana’, helping in suppression of aging, development of positive physical and mental health along with strengthening the immune system and maintenance of youthfulness [1], 34]. WS has been used for thousands of years and its demand has been rising continuously in the nutraceutical market globally. Hence, to meet the international standards of different countries, it is necessary to assess the PK parameters of the standardized WS root extracts. The PK parameters of WS have been less explored thus far and need consideration to understand how the body reacts to individual bioactive components of this plant. Hence, this open label, single dose study was designed to evaluate the PK parameters of WS (not less than 2.5 % of withanolides) root extract capsules (400 mg) in 12 healthy volunteers. Additionally, safety, adverse effects, and tolerability of the WS root extract capsules were also investigated.

Upon administration of WS root extract capsules (400 mg), the individual bioactive components were measured using UHPLC-MS/MS for PK parameters. C max and T max were found to be highest for 12-deoxy-withastramonolide followed by withaferin A. Literature pre-clinical studies have revealed that C max and T max vary from 7.2 ng- 1.8 μM and 0.11–0.75 h in vivo for various constituents present in WS root extract [3], 26], 35], 36]. Alluri et al. [25] studied the comparative PK of 300 mg (Prolanza™) sustained release capsules containing 15 mg withanolides of WS root extract in a randomized, two-treatment, crossover, oral single-dose on 14 healthy men. Results demonstrated that C max for withanolide A and 12-deoxy-withastramonolide was 0.49 ng/mL and 2.67 ng/mL, respectively against the reference (organic KSM-66 Ashwagandha extract vegan capsule); whereas, T max was reported as 1.0 and 2.0 h, respectively, and t1/2 was 7.46 and 7.53 h for withanolides A and 12-deoxy-withastramonolide, respectively.

AUC is a determinant of drug bioavailability and reflects the aggregate drug reaching the systemic circulation. The AUC was found to highest for withaferin A followed by 12-deoxy-withastramonolide in male participants is owing to longer t1/2 compared to other constituents reflecting longer retention of the drug in the body which could be a crucial parameter in optimization of efficacy profile of the drug [37]. Kim et al. [19] highlighted the significance of drug concentration of total withanolides as their group with higher concentration of withanolides (WS-35; 35 % withanolide glycosides) was found to have higher AUC and C max , thus, longer half-life time and absorption compared to WS-2.5 (2.5 % withanolides). Additionally, sustained release formulation of WS could also improve the efficacy of drug by maintaining the constant therapeutic level of the active ingredients in the body [25].

Our study reported the simultaneous determination of four bioactive constituents present in WS root extract; however, previous clinical studies were limited to determination of two or three constituents in WS root extract capsules [25]. Additionally, previous PK studies were focused on single gender (male) whereas, our study included both the genders. Further, safety and tolerability profiles of WS root extract capsules on healthy volunteers were also investigated. Previous literature has revealed a promising safety profile of WS along with its constituents, even at higher doses. In a recent study, Vaidya et al. [29] suggested a safer daily dose of 1,000 mg of standardized WS root extract capsules containing total withanolides with optimal concentration of 7.50 mg. Numerous indications were evaluated in the trial, including adverse event profile, organ function tests, X-ray, ECG, and the tolerance of the WS extract capsules. The end point observations revealed no significant changes in liver, kidney, and thyroid functions. The study concluded that daily dose of 1,000 mg of WS root extract capsule can be safe to use in healthy men without producing any adverse effect in these participants [29].

In another study by Raut et al. [27], ascending doses of WS aqueous extract were administered for 30 days to 18 healthy participants. The dosage regimen comprised of 750 mg/day initial 10 days, followed by 1,000 mg/day succeeding 10 days, followed by 1,250 mg/day for the last 10 days. Further, volunteers were observed for unfavorable consequences via self-reported adverse incidents, several medical tests, viz. fasting sugar, lipid profile, renal, liver function tests, and ECG. No adverse effects were reported except for the one participant who at the lowest dosage showed increased appetite and libido, and hallucinatory effects along with vertigo, which in turn led to withdrawal from the trial. In an open label, randomized, two period, single dose, crossover clinical study, when subjects were given WS sustained release root extract capsules 600 mg (2 capsules), each containing 15 mg withanolides, there were no significant abnormal changes, or adverse effects during and post study in 14 healthy men [25]. Similarly, a long term, randomized, double-blind, placebo-controlled study of sustained release WS root extract with a drug dose (300 mg) daily in 125 participants for 90 days did not show any instance of adverse consequences [38].

In another randomized, placebo-controlled, double-blind safety study including 80 healthy participants (40 males, 40 females), oral administration of WS (300 mg) two times daily for 8 weeks was evaluated for safety parameters. The results did not indicate any adverse consequences and no statistically significant variations or abnormality effects in any of the treated volunteers including thyroid hormonal profile [28]. In a phase-1 trial with patients of high-grade, advanced osteosarcoma, subjects tolerated well a high dose of 4,800 mg which contained 216 mg of withaferin-A per day. Among 11 patients, five exhibited higher liver enzymes at grade 1, and two reported skin rash. Additionally, instances of fever, edema, diarrhea, and fatigue were also reported, although no grade 3 or 4 adverse events were detected [39]. The safety assessment studies of WS indicate that the WS extracts when given in specified dosage are well-tolerated with minimal side effects. Individual reactions can differ, though, and more studies are necessary to fully evaluate its safety, especially in a range of clinical settings and groups [29].

Safety assessment is one of the most crucial components of any clinical investigation. Any formulation, either pharmaceutical or nutraceutical supplements, is subject to safety, tolerability, and efficacy assessments. Plant based products are generally considered to be safe, however, such products after rigorous evaluation contribute in maintaining better human health. In our study, the WS root extract capsules confirmed safety on hematological parameters and biochemical organ functions. No side effects or tolerance were reported along with no substantial effect on critical functions like blood pressure, respiration, and pulse rate during baseline and at the end of the study. Additionally, no clinically considerable modifications were perceived in the participants’ chest X-rays as well as ECGs at the end of the study.

Hepatoprotection has been a primary concern for pharmaceutical and nutraceutical usage. In the present study, no significant alterations were noticed during estimation of the biochemical markers of the liver, which highlights the hepatoprotective effect of WS which was similar to previous literature [28], 29], 40]. The current clinical study also reported no substantial changes in the renal function markers. In total, hematological and biochemical parameters in the present study did not show any clinically significant alterations at p<0.05 along with high tolerability towards the dose. However, this is inconsistent with some previously reported literature studies. Indeed, the safety profile of WS may vary from individual to individual as well as standardized extracts.

In our study, gender-based differences in the PK parameters have been observed. Although there was no significant difference in T max between male and female volunteers, it was observed that the C max and AUC0→t were lower in females than in males, except for WSIV. Furthermore, the t1/2 and MRT in females was lower than in males for all four constituents. In addition, the clearance in females was higher than in males, except for WSIV, whereas the elimination rate constant was higher in females than in males for all four constituents. These changes in PK between male and female volunteers may be attributed to different rates of Phase 1 metabolism of the four constituents of WS by the CYP450 family as well as potential epigenetic modulation of target occupancy. Further studies are needed to elucidate the exact mechanistic basis for these gender-based differences in the PK of WS.

The present study presented PK parameters of WS root extract in male and female volunteers along with scientific and clinical evaluations of safety and tolerability profiles, although the study is limited by its small size and short duration. Therefore, a trial evaluating long term effects of WS in males and females along with vigilant monitoring to understand the interactions with concurrent drugs is needed.

Conclusions

The present study demonstrated the PK properties of bioactive compounds of WS root extract in male and female healthy volunteers. Additionally, safety and tolerability studies suggested safe use of WS root extract at 400 mg daily dose. The hematological and biochemical studies revealed no significant changes and were within the normal limited range at the end of the study. Our PK study will offer insights into future pharmacometric studies involving pharmacodynamic correlation (PK/PD) and biomarker analyses. Future long-term studies in a larger cohort and longer duration are needed to further validate the results of this pilot study.


Corresponding author: Dr. Sujit Nair, MPharm, PhD, Chief Scientific Officer, Phytoveda Pvt. Ltd, Mumbai, India; and Viridis Biopharma Pvt. Ltd., V.N. Purav Marg, Mumbai 400 022, India, E-mail:

Acknowledgments

The authors are thankful to Dr. Vaishnavi Patil (Mprex Healthcare Pvt. Ltd., Pune-411057, Maharashtra, India) for her support in the clinical trial of WS root extract capsules.

  1. Research ethics: The study adhered to the Declaration of Helsinki and was approved by the Institutional Ethical Committee of Lokmanya Medical Research Centre, Chinchwad, Pune, Maharashtra, India (ECR/175/Inst/MH/2013/RR-19).

  2. Informed consent: Informed consent was acquired from all participants included in this study.

  3. Author contributions: Eshita Sharma: Writing – original draft, Writing – review & editing; Gayatri Ganu: Methodology, Project administration, Resources; Ketan Kshirsagar: Conceptualization, Investigation, Supervision, Validation; Ashwin Shah: Writing – review & editing; Umakant Mahale: Writing – review & editing; Anirudh Mehta: Conceptualization, Supervision; Sujit Nair: Conceptualization, Supervision, Formal analysis, Software, Writing – review & editing. All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  4. Use of Large Language Models, AI and Machine Learning Tools: None declared.

  5. Conflict of interest: LongeFera™ is marketed by Phytoveda Pvt. Ltd., India and Phytoveda, USA. ES, AM and SN are employees of Phytoveda Pvt. Ltd., Mumbai, India and Viridis Biopharma Pvt. Ltd., Mumbai, India. However, these authors had no role in the conduct of the clinical trial which was outsourced to an external Contract Research Organisation (CRO), i.e., Mprex Healthcare Pvt. Ltd., Pune, India. GG is an employee of Mprex Healthcare Pvt. Ltd., India, and declares no conflict of interest. KK is the clinician investigator who conducted the study at the hospital site and declares no conflict of interest. All other authors state no conflict of interest.

  6. Research funding: The study was funded by Phytoveda Pvt. Ltd., Mumbai, India.

  7. Data availability: Data will be made available from the corresponding author on reasonable request.

References

1. Basudkar, V, Gujrati, G, Ajgaonkar, S, Gandhi, M, Mehta, D, Nair, S. Emerging vistas for the nutraceutical Withania somnifera in inflammaging. Pharmaceuticals 2024;17:597. https://doi.org/10.3390/ph17050597.Suche in Google Scholar PubMed PubMed Central

2. KrishnaRaju, AV, Somepalli, V, Thanawala, S, Shah, R. Efficacy and anti-inflammatory activity of Ashwagandha sustained-release formulation on depression and anxiety induced by chronic unpredictable stress: in vivo and in vitro studies. J Exp Pharmacol 2023;15:291–305. https://doi.org/10.2147/JEP.S407906.Suche in Google Scholar PubMed PubMed Central

3. Modi, SJ, Tiwari, A, Ghule, C, Pawar, S, Saste, G, Jagtap, S, et al.. Pharmacokinetic study of withanosides and withanolides from Withania somnifera using ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). Molecules 2022;27:1476. https://doi.org/10.3390/molecules27051476.Suche in Google Scholar PubMed PubMed Central

4. Wijeratne, EMK, Xu, YM, Scherz-Shouval, R, Marron, MT, Rocha, DD, Liu, MX, et al.. Structure-activity relationships for withanolides as inducers of the cellular heat-shock response. J Med Chem 2014;57:2851–63. https://doi.org/10.1021/jm401279n.Suche in Google Scholar PubMed

5. Baker, C, Kirby, JB, O’Connor, J, Lindsay, KG, Hutchins, A, Harris, M. The perceived impact of Ashwagandha on stress, sleep quality, energy, and mental clarity for college students: qualitative analysis of a double-blind randomized control trial. J Med Food 2022;25:1095–101. https://doi.org/10.1089/jmf.2022.0042.Suche in Google Scholar PubMed

6. Das, R, Rauf, A, Akhter, S, Islam, MN, Emran, TB, Mitra, S, et al.. Role of withaferin A and its derivatives in the management of Alzheimer’s disease: recent trends and future perspectives. Molecules 2021;26:3696. https://doi.org/10.3390/molecules26123696.Suche in Google Scholar PubMed PubMed Central

7. Deshpande, A, Irani, N, Balkrishnan, R, Benny, IR. A randomized, double blind, placebo controlled study to evaluate the effects of Ashwagandha (Withania somnifera) extract on sleep quality in healthy adults. Sleep Med 2020;72:28–36. https://doi.org/10.1016/j.sleep.2020.03.012.Suche in Google Scholar PubMed

8. Haque, IM, Mishra, A, Kalra, BS, Chawla, S. Role of standardized plant extracts in controlling alcohol withdrawal syndrome-an experimental study. Brain Sci 2021;11:919. https://doi.org/10.3390/brainsci11070919.Suche in Google Scholar PubMed PubMed Central

9. Mehta, V, Chander, H, Munshi, A. Mechanisms of anti-tumor activity of Withania somnifera (Ashwagandha). Nutr Cancer 2021;73:914–26. https://doi.org/10.1080/01635581.2020.1778746.Suche in Google Scholar PubMed

10. Sharma, PK, Kumar, L, Goswami, Y, Pujani, M, Dikshit, M, Tandon, R. The aqueous root extract of Withania somnifera ameliorates LPS-induced inflammatory changes in the in vitro cell-based and mice models of inflammation. Front Pharmacol 2023;14:1139654. https://doi.org/10.3389/fphar.2023.1139654.Suche in Google Scholar PubMed PubMed Central

11. Singh, M, Jayant, K, Singh, D, Bhutani, S, Poddar, NK, Chaudhary, AA, et al.. Withania somnifera (L.) Dunal (Ashwagandha) for the possible therapeutics and clinical management of SARS-CoV-2 infection: plant-based drug discovery and targeted therapy. Front Cell Infect Microbiol 2022;12:933824. https://doi.org/10.3389/fcimb.2022.933824.Suche in Google Scholar PubMed PubMed Central

12. Singh, N, Bhalla, M, de Jager, P, Gilca, M. An overview on Ashwagandha: a Rasayana (rejuvenator) of Ayurveda. Afr J Tradit Complement Altern Med 2011;8:208–13. https://doi.org/10.4314/ajtcam.v8i5S.9.Suche in Google Scholar PubMed PubMed Central

13. Tiwari, S, Gupta, SK, Pathak, AK. A double-blind, randomized, placebo-controlled trial on the effect of Ashwagandha (Withania somnifera dunal.) root extract in improving cardiorespiratory endurance and recovery in healthy athletic adults. J Ethnopharmacol 2021;272:113929. https://doi.org/10.1016/j.jep.2021.113929.Suche in Google Scholar PubMed

14. Kushwaha, S, Soni, VK, Singh, PK, Bano, N, Kumar, A, Sangwan, RS, et al.. Withania somnifera chemotypes NMITLI 101R, NMITLI 118R, NMITLI 128R and withaferin A protect Mastomys coucha from Brugia malayi infection. Parasite Immunol 2012;34:199–209. https://doi.org/10.1111/j.1365-3024.2012.01352.x.Suche in Google Scholar PubMed

15. Dutta, R, Khalil, R, Green, R, Mohapatra, SS, Mohapatra, S. Withania somnifera (Ashwagandha) and withaferin A: potential in integrative oncology. Int J Mol Sci 2019;20:5310. https://doi.org/10.3390/ijms20215310.Suche in Google Scholar PubMed PubMed Central

16. Wongtrakul, J, Thongtan, T, Kumrapich, B, Saisawang, C, Ketterman, AJ. Neuroprotective effects of Withania somnifera in the SH-SY5Y Parkinson cell model. Heliyon 2021;7:e08172. https://doi.org/10.1016/j.heliyon.2021.e08172.Suche in Google Scholar PubMed PubMed Central

17. Guo, S, Rezaei, MJ. The benefits of Ashwagandha (Withania somnifera) supplements on brain function and sports performance. Front Nutr 2024;11:1439294. https://doi.org/10.3389/fnut.2024.1439294.Suche in Google Scholar PubMed PubMed Central

18. Remenapp, A, Coyle, K, Orange, T, Lynch, T, Hooper, D, Hooper, S, et al.. Efficacy of Withania somnifera supplementation on adult’s cognition and mood. J Ayurveda Integr Med 2022;13:100510. https://doi.org/10.1016/j.jaim.2021.08.003.Suche in Google Scholar PubMed PubMed Central

19. Kim, S-K, Venkatesan, J, Rathi, P, Antony, B. Pharmacokinetics and bioequivalence of Withania somnifera (Ashwagandha) extracts - a double blind, crossover study in healthy adults. Heliyon 2023;9:e22843. https://doi.org/10.1016/j.heliyon.2023.e22843.Suche in Google Scholar PubMed PubMed Central

20. Nair, S, Kong, A-NT. Emerging roles for clinical pharmacometrics in cancer precision medicine. Curr Pharmacol Rep 2018;4:276–83. https://doi.org/10.1007/s40495-018-0139-0.Suche in Google Scholar PubMed PubMed Central

21. Shah, D, Gandhi, M, Kumar, A, Cruz-Martins, N, Sharma, R, Nair, S. Current insights into epigenetics, noncoding RNA interactome and clinical pharmacokinetics of dietary polyphenols in cancer chemoprevention. Crit Rev Food Sci Nutr 2023;63:1755–91. https://doi.org/10.1080/10408398.2021.1968786.Suche in Google Scholar PubMed

22. Nair, S, Kong, A-HT. Pharmacometrics of nutraceutical sulforaphane and its implications in prostate cancer prevention. J Chin Pharmaceut Sci 2016;25:12–22. https://doi.org/10.5246/jcps.2016.01.002.Suche in Google Scholar

23. Nair, S, Iyer, A, Vijay, V, Bandlamudi, S, Llerena, A. Pharmacokinetics and systems pharmacology of monoclonal antibody olaratumab for inoperable soft tissue sarcoma. Adv Mod Oncol Res 2017;3. https://doi.org/10.18282/amor.v3.i3.194.Suche in Google Scholar

24. Nair, S. Pharmacometrics and systems pharmacology of immune checkpoint inhibitor nivolumab in cancer translational medicine. Adv Mod Oncol Res 2016;2:18. https://doi.org/10.18282/amor.v2.i1.46.Suche in Google Scholar

25. Alluri, VKR, Thanawala, S, Upadhyay, V. A comparative pharmacokinetics study of Ashwagandha (Withania somnifera) root extract sustained-release capsules: an open-label, randomized, two treatment, two-sequence, two period, single-dose crossover clinical study. Int J Basic Clin Pharmacol 2022;11:26–34. https://doi.org/10.18203/2319-2003.ijbcp20214831.Suche in Google Scholar

26. Patil, D, Gautam, M, Mishra, S, Karupothula, S, Gairola, S, Jadhav, S, et al.. Determination of withaferin A and withanolide A in mice plasma using high-performance liquid chromatography-tandem mass spectrometry: application to pharmacokinetics after oral administration of Withania somnifera aqueous extract. J Pharm Biomed Anal 2013;80:203–12. https://doi.org/10.1016/j.jpba.2013.03.001.Suche in Google Scholar PubMed

27. Raut, AA, Rege, NN, Tadvi, FM, Solanki, PV, Kene, KR, Shirolkar, SG, et al.. Exploratory study to evaluate tolerability, safety, and activity of Ashwagandha (Withania somnifera) in healthy volunteers. J Ayurveda Integr Med 2012;3:111–4. https://doi.org/10.4103/0975-9476.100168.Suche in Google Scholar PubMed PubMed Central

28. Verma, N, Gupta, SK, Tiwari, S, Mishra, AK. Safety of Ashwagandha root extract: a randomized, placebo-controlled, study in healthy volunteers. Complement Ther Med 2021;57:102642. https://doi.org/10.1016/j.ctim.2020.102642.Suche in Google Scholar PubMed

29. Vaidya, VG, Gothwad, A, Ganu, G, Girme, A, Modi, SJ, Hingorani, L. Clinical safety and tolerability evaluation of Withania somnifera (L.) Dunal (Ashwagandha) root extract in healthy human volunteers. J Ayurveda Integr Med 2024;15:100859. https://doi.org/10.1016/j.jaim.2023.100859.Suche in Google Scholar PubMed PubMed Central

30. Wangikar, P, Chaudhari, P, Sharma, E, Godse, C, Vora, A, Nair, S. Acute and sub-chronic oral GLP toxicity of Withania somnifera root extract in Sprague Dawley Rats. Drug Metabol Pers Therapy 2024;39:145–58. https://doi.org/10.1515/dmpt-2024-0056.Suche in Google Scholar PubMed

31. Sharma, AK, Basu, I, Singh, S. Efficacy and safety of Ashwagandha root extract in subclinical hypothyroid patients: a double-blind, randomized placebo-controlled trial. J Altern Complement Med 2018;24:243–8. https://doi.org/10.1089/acm.2017.0183.Suche in Google Scholar PubMed

32. Chandrasekhar, K, Kapoor, J, Anishetty, S. A prospective, randomized double-blind, placebo-controlled study of safety and efficacy of a high-concentration full-spectrum extract of Ashwagandha root in reducing stress and anxiety in adults. Indian J Psychol Med 2012;34:255–62. https://doi.org/10.4103/0253-7176.106022.Suche in Google Scholar PubMed PubMed Central

33. Vaidya, VG, Naik, NN, Ganu, G, Parmar, V, Jagtap, S, Saste, G, et al.. Clinical pharmacokinetic evaluation of Withania somnifera (L.) Dunal root extract in healthy human volunteers: a non-randomized, single dose study utilizing UHPLC-MS/MS analysis. J Ethnopharmacol 2024;322:117603. https://doi.org/10.1016/j.jep.2023.117603.Suche in Google Scholar PubMed

34. Saha, P, Ajgaonkar, S, Maniar, D, Sahare, S, Mehta, D, Nair, S. Current insights into transcriptional role(s) for the nutraceutical Withania somnifera in inflammation and aging. Front Nutr 2024;11. https://doi.org/10.3389/fnut.2024.1370951.Suche in Google Scholar PubMed PubMed Central

35. Dai, T, Jiang, W, Guo, Z, Wang, Z, Huang, M, Zhong, G, et al.. Studies on oral bioavailability and first-pass metabolism of withaferin A in rats using LC-MS/MS and Q-TRAP. Biomed Chromatogr 2019;33:e4573. https://doi.org/10.1002/bmc.4573.Suche in Google Scholar PubMed

36. Thaiparambil, JT, Bender, L, Ganesh, T, Kline, E, Patel, P, Liu, Y, et al.. Withaferin A inhibits breast cancer invasion and metastasis at sub-cytotoxic doses by inducing vimentin disassembly and serine 56 phosphorylation. Int J Cancer 2011;129:2744–55. https://doi.org/10.1002/ijc.25938.Suche in Google Scholar PubMed

37. Czock, D, Scholle, C, Rasche, FM, Schaarschmidt, D, Keller, F. Pharmacokinetics of valganciclovir and ganciclovir in renal impairment. Clin Pharmacol Ther 2002;72:142–50. https://doi.org/10.1067/mcp.2002.126306.Suche in Google Scholar PubMed

38. Gopukumar, K, Thanawala, S, Somepalli, V, Rao, TSS, Thamatam, VB, Chauhan, S. Efficacy and safety of Ashwagandha root extract on cognitive functions in healthy, stressed adults: a randomized, double-blind, placebo-controlled study. Evid Based Complement Alternat Med 2021;2021:8254344. https://doi.org/10.1155/2021/8254344.Suche in Google Scholar PubMed PubMed Central

39. Pires, N, Gota, V, Gulia, A, Hingorani, L, Agarwal, M, Puri, A. Safety and pharmacokinetics of Withaferin-A in advanced stage high grade osteosarcoma: A phase I trial. J Ayurveda Integr Med 2020;11:68–72. https://doi.org/10.1016/j.jaim.2018.12.008.Suche in Google Scholar PubMed PubMed Central

40. Sultana, R, Sultan, Z. In vitro effect of Withania somnifera, Mucuna pruriens and Pausinystalia johimbe on hepatic cytochrome P450 in rat. Bangladesh Pharm J 2018;21:118. https://doi.org/10.3329/bpj.v21i2.37922.Suche in Google Scholar


Supplementary Material

This article contains supplementary material (https://doi.org/10.1515/dmpt-2024-0089).


Received: 2024-11-08
Accepted: 2025-01-16
Published Online: 2025-02-19

© 2025 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Heruntergeladen am 14.9.2025 von https://www.degruyterbrill.com/document/doi/10.1515/dmpt-2024-0089/html
Button zum nach oben scrollen