Subscribe to RSS
DOI: 10.1055/a-2686-6315
Factors Influencing Clinical Trials of Herbal Medicinal Products – Using Ginger as Example
Authors
Abstract
Ginger (Zingiber officinale) has a global use as a spice, an ingredient of beverages, food supplements (syn. dietary supplements), as well as herbal medicinal products. Since the last update of ginger in ESCOP Monographs in 2009 a significant number of papers concerning its bioactive constituents and clinical uses have been published. From this large number and selecting those references (almost 500) considered most relevant to clinical aspects and therapeutic indications, the following issues are considered to be potentially important to research on other medicinal plants: [i] quality assessment; [ii] pre-clinical (in vivo) studies; [iii] quality of clinical trials; [iv] ethnicity of clinical trial participants and [v] effects of sex-gender on activity and therapeutic indications.
Introduction
Herbal medicinal products [HMPs] are particularly suitable for treating non-life-threatening ailments, and the use of plants or herbs to manage health conditions [phytotherapy] has become increasingly popular and commercially important in recent years. However, a number of aspects have arisen regarding the quality and scientific assessment of these herbal products [1], [2], [3]. International (such as the European Medicines Agency; EMA) and national regulatory bodies have the responsibility of monitoring the quality of medicinal products including herbal ones. Like for all other medicines, their controlled quality is mandatory, but – apart from those products on tradition only – clinical data to assess their safety and efficacy are required. Many aspects of publications concerning human clinical trials with herbal materials are too variable, with reviews and analyses often concluding “that problems exist regarding the quality of the material sampled, the small sizes of the trials and poor evaluation systems or parameters employed to reach conclusions”. Also, scientific evidence is often limited and needs to be improved to allow integration into guidelines and standard regimens of evidence-based clinical practice [4], [5], [6].
Search Strategy
The second edition of ESCOP Monographs was published in 2003 and included Zingiberis Rhizoma with 58 references. Another version (updated by I. Hook) followed in 2009 with 147 references. To bring the information up to date, the CAS SciFinder from 2007 to 2022 was searched with the words “Zingiber officinale, ginger, rhizome”. This resulted in a potential 34 004 references. To rationalise this large number, the additional terms then applied were “activity, bioactivity, clinical trials”, resulting in 1,485 references. Finally the process was repeated for 2023 when a further total 1012 references were reported, which were then rationalised again with the terms “bioactivity, clinical trials”, resulting in a further 217 relevant references. Abstracts of all these were examined for methodology and relevance resulting in a total of 1701 publications, which on further scrutiny, resulted in many in vitro trials being excluded. Full publications were always examined for greater details, such as ginger product and dose information, animal and human genders, toxicity, etc.
Quality Assessment
Constituents
A challenge with herbal medicines is the large number of constituents with potential bioactivity versus conventional drugs with usually only one or two biologically active constituents [7], [8]. However, as with all medicines, the ultimate demonstration of therapeutic usefulness is clinical trials for safety and efficacy, but these can only be scientifically evaluated when the herbal material being used is authenticated, standardized and quality controlled. The active constituents of ginger are considered to be a series of pungent polyphenolic homologues of gingerol (mainly 6-gingerol) [9] and shogaols, present in the oleoresin which makes up 4 to 7.5% of the dried rhizome material. Of this oleoresin, 25% are the pungent principles; i.e. circa 1 to 2%. Also present is an essential oil (0.25 to 5% v/w) made up predominantly of sesquiterpenes [10] such as a-zingiberene (17 to 37%).
The regulation of herbal medicines varies greatly between countries and global regions [11], but national Pharmacopoeial monographs defining the expected quality are published. However, what is expected by way of quality is variable. For example in the Chinese Pharmacopoeia [12] the quality of dried ginger is determined by the essential oil content (not less than 0.8%) and 6-gingerol (not less than 0.6%) analysed by HPLC. In the European Pharmacopoeia [13], the quality is assessed by the amount of essential oil (not less than 1.5%) with constituent identification by (HP)TLC. For comparison the USP-NF states that ginger should contain not less than 1.8% of essential oil, not less than 0.8% of gingerols and gingerdiones and not more than 0.18% of shogaols as analysed by TLC and HPLC [14]. Marketing authorization and licencing of these herbal materials is also ambiguous with three categories potentially classified as HMPs being available on the European market: (i) HMPs (ii) food supplements or (iii) cosmetics [15]. According to the EMA/HMPC document [16] there are currently 13 officially licensed products containing ginger in the EU [Germany, Lithuania, Netherlands, Poland each with × 1, Austria with × 3 and UK with × 5]. However, a market survey carried out in 2022 found that in addition, many foods and food supplements have appeared on the market over the last ten years, with demand expected to grow in the coming years [17]. In 2021 the UK FSA Exposure team examined information on ginger supplements and food intake in women of childbearing age to assess the potential of its constituents and possible contaminants (including heavy metals) on reproductive toxicity, on a long-term basis. They listed × 18 supplements, × 7 foods and × 21 juices/drinks/teas [18].
In the United States ginger products are marketed as dietary supplements, not as herbal medicinal products, and neither the FDA nor any other federal or state agency routinely tests these supplements for quality, prior to sale. A recent market survey of sales in the United States showed ginger to be one of the most widely consumed herbal supplements [19] and a search by Husain [20] in the NIHʼs Dietary Supplement Label Database resulted in more than 3500 ginger-based products sold via major retailers. Products are highly variable in quality. Schwerter [21] for example found that 6-gingerol concentration of the ginger powder of dietary supplements ranged from 0.0 to 9.43 mg/g, 6-shogaol from 0.16 to 2.18 mg/g, 8-gingerol from 0.00 to 1.1 mg/g and 10-gingerol from 0.00 to 1.40 mg/g. Similarly a quality assessment in 2022 by an independent company [22] found that of 18 products, 7 contained low levels of active constituents and nearly all had shogaol levels higher than USP specifications. In many clinical trials the content of actives is not determined or published.
Pesticides
In 2022 the annual world production of ginger was approximately 4.1 million metric tons, with China, India and Nigeria among major producers. It has long crop-growing periods in the fields (210 to 240 days after planting) thereby requiring intensive use of various pesticides [23]. After harvesting, ginger processing can include washing, peeling, boiling, drying and powdering, before solvent extraction or distillation occurs. Each process varies in how the levels of pesticides resulting in the ginger end-product are affected. Since ginger is also commonly used for cooking or as a dietary supplement, a risk for exposure to pesticides exists from frequent or excessive consumption, resulting in pesticide residue analysis of ginger being required by many countries [23]. Evaluation of more than 3 million sets of data on residues in herbal drugs available to European manufacturers, found that in most cases (98%) the limits of quantitation were lower or equal to 0.05 mg/kg (0.05 ppm), with 63% lower or equal to 0.01 mg/kg, indicating that the Ph.Eur. limits on pesticide residues were not exceeded [24].
Heavy metals
Apart from pesticide residues Pharmacopoeial monographs specify, as part of quality, limits on microbial contaminants and heavy metals (arsenic, cadmium, chromium, cobalt, lead, mercury, nickel, vanadium). These purity determinations are rarely carried out or results published. Such heavy metal contamination above accepted threshold concentrations in herbal medicines is presently a global threat to human health. A recent analysis of 1773 samples from around the world of 86 different types of commonly used Chinese herbal medicines found 30.5% with at least one, over the accepted Chinese Pharmacopoeia limit [25]. Also, Danggui (Radix Angelica sinensis) commonly consumed by pregnant and nursing mothers in Malaysia, was found on sample analysis to contain high concentrations of heavy metals, posing a potential health risk of transfer from mother to foetus and infant [26]. Heavy metal content of soil and root accumulation is a known cause of abiotic stress affecting plant health, annual growth, enzyme activity affecting bioactive metabolite contents (e.g. ginsenosides in Panax ginseng C. A. Meyer) [27]. Some medicinal plants are also active hyper-accumulators of heavy metals from contaminated soil, for example St. Johnʼs Wort (Hypericum perforatum L.) and Chamomile (Matricaria recutita L.) can amass a relatively high concentration of cadmium ranging from 1 to 4% of biomass [28], [29].
Ginger also can accumulate toxic levels of mercury from polluted soil [30] and the recent quality assessment in the US [22] found 0.53 mcg of lead per g of a product, exceeding the daily allowance and indicative of “reproductive harm”. Analysis of ginger from Ethiopia for heavy metals found mean concentrations (mg/kg) ranging from 4.63 to 5.43 for cadmium, 2.17 to 4.44 for chromium, 62.52 to 65.14 for copper, 77.71 to 81.12 for iron, 6.49 to 7.58 for nickel and 16.74 to 19.31 for zinc, but lead was not detected [30]. There is therefore a ʼpotentialʼ for ginger products when taken during pregnancy and lactation to have reprotoxic effects at high supplemental doses. For heavy metals the Ph. Eur. general monograph Herbal drugs [13] has the following limits: cadmium 1.0 ppm, lead 5.0 ppm, mercury 0.1 ppm. According to EMA/HMPC/765 220/2022 [16] in EU/EEA member states there are currently 13 medicinal ginger products of Ph. Eur. quality marketed as well as many foods and food supplements of varying quality [18]. Although pesticide residues [24] or heavy metal contamination in products in general appear not to be a major problem, analysis of ginger rhizome samples (x6) for contents of heavy metals found cadmium and lead levels to be within limits, but 2 out of the 6 samples had mercury above the permitted limit [31]. Overall the Ph. Eur. limits set for cadmium, lead and mercury are considered appropriate and samples from organic production have revealed a lower occurrence of these elements as compared to samples from conventional production. However, compared to the period 2008 – 2015, the results from the current updated period (2016 – 2021) show significantly more positive findings for heavy metal elements in herbal drugs and essential oils [32].
Pre-clinical (in vivo) animal studies
The use of animals in testing of herbal medicines, while offering undeniable advantages, remains a subject of intense debate and contention, mainly arising from the ethical challenges it presents, the substantial financial burden it imposes, as well as its inability to reliably predict human responses [33]. To overcome this, newer approaches (e.g. systems pharmacology) are being applied to explore pharmacological effects of multi-component herbal medicine [34] and help to identify and prevent the risk of herb–drug interactions at an early stage of the drug development process [35].
Although sampling animals of both sexes and of various hormonal states has produced new discoveries that influence drug development and patient care, and countries have typically enacted legislation that requires sex/gender diversity in sponsored human studies, these guidelines are rarely enforced to studies on animals. Much basic research still uses animal models that focus on males and excludes females [36], creating a sex bias in biomedical studies that needs to be minimised [37].
The use of appropriate animal models to assess a possible effect in humans is invaluable and rodents are the most frequently used, with mice and rats making up approximately 95% of all laboratory animals. However, for example in diabetes mellitus research, for T1D Yagihashi [38] lists six strains of rats + mice and for T2D, four types of rat and eight types of mice. Overall this type of research is poorly standardised, as not only is model type important, but also age, sex, genetic background and husbandry practices, all of which can impact the results [39]. For example, in the Morris water maze test using ICR mice, females showed superior learning to, and greater memory abilities than, males [40], an effect however that was not found using C57BL/6 mice [41]. Female mice are often excluded from animal studies but their inclusion is important to improve detection of real drug effects [42]. This gender bias creates three problems: [i] less knowledge about disease processes in females due to underutilization of female animals, [ii] an inability to use sex as a variable, e.g. in regulation of immune function and [iii] missed female-specific phenomena such as pregnancy and menopause. Data extracted from drug dossiers submitted to the EMA for marketing authorisation between 2011 and 2015, showed female animals were included in only 9% of the pharmacodynamics studies, although both female and male animals were included in all toxicology studies [43]. Also, for drugs in general there is a “poor translation rate (over 90%) from animal results to human studies” [44] and often with a citation bias on the positive with under-citation of the negative [45].
Conventional medicines are known to affect male and female human patients differently and this has now occasionally been reported in animals when treated with herbal extracts. For example, with a St. Johnʼs Wort (H. perforatum L.) extract, hypericin plasma levels were significantly more elevated in female three-month-old mice than in males [46], while in rats cognitive impairment was found, possibly through a reduction in the levels of neurotrophic factors, an effect that was expressed more in females than in males [47]. In Centella (Centella asiatica (L.) Urban) an extract was found to have different effects in the memory retention of male and female C57BI/6 mice [48]. Similarly when 5 xFAD mice (a model of Alzheimerʼs disease) were given this C. asiatica extract, results showed that after 4.5 weeks, in female mice there was no loss in the conditioned fear response, but this was diminished in male mice [49]. To better understand the effects of gender and how varying concentrations of C. asiatica aqueous extract affects the brain differently, a metabolomic analysis of cortical tissue collected from male and female 5 xFAD mice and wild-type (WT) litter mates was carried out. With a dose of 500 mg/kg/d, for example, in 5 xFAD females nine pathways were altered significantly compared to seven in 5 xFAD males and six pathways in WT females compared to four in WT males [50]. With Rosemary (Salvia rosmarinus Spenn.) a dose of 500 mg/kg resulted in a significant decline in spermatogenesis of male rats attributed to a significant decrease in testosterone, while in female rats it led to reduced fertility due to an increased number of foetal resorptions [51]. In ginger different doses (200 mg/kg for males and 250 mg/kg for females) were found to modulate gamma radiation-induced conditioned taste aversion in rats in a gender-specific manner due to hormonal fluctuations [52].
Examination of the references for the ESCOP Zingiberis Rhizoma Monograph update, the following was found: in vivo animal studies cited a total of 87 trials of which 74 used rats or mice. Rats were used in 41 trials with the following use of genders: male × 29, female × 1, both sexes × 7 or no gender given × 4. Mice were used in 33 trials with the genders as follows: male × 21, female × 6, both sexes × 1 or no gender × 4; i.e. where gender was given 77% were male. Rarely were results for the genders analysed separately and/or results given. Also, the vast majority of results showed a positive effect after ginger treatment, which could suggest a possible selection bias for studies that could confirm the therapeutic indications of ginger. Similarly in the narrative review by Crichton [53] on mechanisms of ginger actions of 36 in vivo animal studies, mainly mice and rats were used and no gender was given in 36%, but where gender was given 87% were male.
Clinical Trials
In a recent systematic review of all the published 109 clinical trials with ginger [54], critical evaluation showed that only 43 met the criterion of having a “high quality of evidence”; i.e. 60% are poorer in quality. Similarly in the umbrella review by Crichton [55] of 22 trials, only 4 (18%) were considered to be of “high quality”. In this study, 133 clinical trials were included (see [Table 1]). Some variables in clinical trials are highlighted below.
|
Indication |
No. of cited studies |
No. of studies with positive outcome |
|---|---|---|
|
a with a response to ginger of 80 to 100%; b with a response to ginger of 60 to 80%; c with a response to ginger of less than 60% |
||
|
Gastrointestinal effects |
||
|
Motility, dyspepsia, salivary flow |
7 |
7 a |
|
Early pregnancy nausea & vomiting (NVP) |
15 |
12 a |
|
Postoperative nausea & vomiting (PONV) |
18 |
13 b |
|
Nausea & vomiting of motion |
14 |
8 c |
|
Chemotherapy nausea & vomiting |
19 |
10 c |
|
Other antiemetic effects |
5 |
5 |
|
Anti-inflammatory and analgesic effects |
||
|
Arthritis (Osteo- & Rheumatoid) |
8 |
7 a |
|
Dysmenorrhoea |
8 |
5 b |
|
Migraine |
3 |
2 b |
|
Metabolic syndrome |
||
|
Weight management |
3 |
3 a |
|
Blood lipids |
8 |
5 b |
|
Glycaemic (insulin) control |
9 |
7 b |
|
Other clinical effects |
16 |
|
Participant ethnicity, dietary habits, pharmacokinetics
The analysis of ginger clinical trials by Anh [54] showed that they took place in the following countries: Iran (× 24), the US (× 13), Australia (× 5), Europe (× 4), Thailand (× 3) and one each in Brazil, China, India and Israel. Data suggests that ethnicity, dietary habits and geographical provenance are closely interlinked and their interrelationship is a key player in determining gut microbiome diversity [56]. Food group intakes – particularly fruit, vegetables and fish – and diet quality scores differ between ethnicities [57], [58] and influences gut microbial composition (microbiome) [59], [60], [61], [62], [63]. For example the infant gut microbiome arises after three months of age and persists through childhood, a variation that is associated with race and ethnicity [64]. South Asian infants were found to have a higher abundance of lactic acid bacteria, while Caucasian infants had higher Clostridiales genera attributed to differences in maternal and infant diets in the first year of life [65]. A large-scale analysis of gut microbiota diversity across US ethnicities (African American, Asian American and Pacific Islander, Caucasian, Hispanic) showed recurrent associations between specific taxa and ethnicity [59]. A two-week intake of ginger powder by healthy American participants (71% female) also resulted in altered aspects of gastrointestinal bacteria composition [66], while a six-week ingestion of ginger by subjects with prior colorectal adenoma, had a limited, but real effect on the faecal microbiome [67]. In healthy Chinese (63 male:60 female) participants a short-term intake of ginger juice was found to have substantial effects on the composition and function of gut microbiota. Significant differences were identified in bacterial genera (× 19) between the control group (women) and ginger group (women) and 15 significant differences at the genus level between the control group (men) and ginger group (men), indicating the importance of analysing both male and females separately [68].
The effects of race or ethnicity on the pharmacokinetics of conventional Western medicines are well recognised [69], but less so for herbal medicines. However, examination of the pharmacokinetics of a multicomponent Chinese herbal preparation (Kang 601) found significant pharmacokinetic differences between healthy male African and Chinese volunteers [70]. With ginger, pharmacokinetic studies have shown that concentrations of actives peaked at circa 1 hr after oral administration when given to healthy volunteers (Americans; gender not specified) [71]. Similarly in mostly Caucasian participants (American; 9 M:22F) tmax for 10-gingerol was at 75 mins and for 6-shogaol at 66 mins with half-lives of the main constituents and their metabolites of 1 – 3 h [72], compared to tmax for 10-gingerol at 38 mins and for 6-shogaol at 30 mins in healthy Indonesian participants (13 M:8F) with half-lives of the metabolites of these constituents at 2.5 – 5.6 h [73].
It is also important to remember that differences in pharmacokinetic effects exist between whole-plant extracts versus pure constituents. In rodent malaria models it was found that interactions occurred between constituents of Sweet Wormwood (Artemisia annua L.) tea or dried whole plant, so that its artemisinin content was more rapidly absorbed than the pure drug and thereby reducing resistance [74], [75]. With ginger similarly a whole-plant extract had different absorption profiles in male mice and enhanced bioavailability compared to single compounds [76]. Also, a study in rats demonstrated that plasma levels of madecassoside, and to a lesser extent asiaticoside, were higher after administration of a standardised extract of C. asiatica than for the pure compounds [77].
Effects of sex-gender on activity and therapeutic indications
Countries typically have enacted legislation that requires inclusion of women in government-sponsored human studies. For example, the U. S. National Institutes of Health require “that women and members of minorities and their subpopulations be included in all human subjects research. Nevertheless sufficient representation of women to allow for sex analysis is required only for Phase III clinical trials” [78]. Data from dossiers submitted to the EMA between 2011 and 2015 [43] showed that inclusion of women in Phase III clinical trials was variable and they were underrepresented for certain diseases. Sex differences in metabolism (Phase I and II) are believed to be the major cause of differential pharmacokinetics. Many CYP450 enzymes (Phase I metabolism) show a sex-dependent difference in activity. Most of Phase II enzymes have a higher activity in men than in women [79]. Sex differences in efficacy are observed with some drugs [43]. Also, the role of gender as a biological factor in the generation of adverse drug reactions (ADRs) is poorly understood with women experiencing almost twice as many ADRs as men. Since most approved clinical trials are conducted on men, it suggests that women could be over-medicated leading to this greater incidence of ADRs [36]. Gender-specific medicine “needs to focus the attention and efforts of the scientific community on understanding the differences of pathophysiology, clinical signs, prevention and treatment of diseases equally represented in men and women” [80], [81], [82].
It is also recognised that journals inconsistently publish sex disaggregated data on drug efficacy [83], although it is known that males and females differ in pharmacological responses to many drug treatments [84], [85], [86]. Examples include motion (travel) sickness, which is more common in children than in adults, more common in women (particularly during menstruation or pregnancy) than in men and not in simulator sickness (e.g. viewing a rotating optokinetic drum) [87]. Ethnic groups differ in how they experience car sickness, with the highest incidence reported in China and the lowest in Germany [88]. Asian women compared to European American and African American women were shown to have a hypersensitivity to motion sickness, while a second study yielded similar results in American-born children of Asian parents [89], [90]. Different ethnic groups also differ in their sensitivity to pain [91], [92] as in post-operative pain and nausea (PONV) [93], [94]. Women of African ethnicity have been shown to have a decreased incidence of PONV compared to Caucasian [95]. Also, pain tolerance was found to be different in Asian as compared to white women [96]. In general women are more susceptible to pain than men [97] and women perceive more pain than men as in pain scores [98], [99]. Migraines too are three to four times more common in women than men [100] and the number of women experiencing osteoarthritis is higher than in men [101]. Women also react differently to drug treatments such as antiemetics [102], [103], anaesthetics [104] and experience different side effects [105]. With depression there are gender-specific mechanisms in activating symptoms and depressive states [106], [107] and womenʼs psychotic symptoms respond to antipsychotic drugs at doses lower than menʼs [108]. Other gender-specific effects include immune responses [109], inflammation [110] and infectious diseases [111].
With medicinal plants it is of interest to note the effect of gender and ethnicity for preferences of essential oils [112]. For example, in healthy school-aged children, male Latinos were more likely to describe Peppermint (Mentha x piperta L.) as “energetic” than male Caucasians, while female Latinos more likely than Caucasian females found Sweet Orange (Citrus x aurantium f. aurantium) “calming”. Effects of ethnicity and gender in odour perception and olfactory evaluation are known to be complex but well-referenced in Chen [113]. Campesi [114] highlighted how gender-sex differentially can influence activities of plant phenols in herbal medicines during critical periods of womenʼs lives. Ingestion of C. asiatica attenuated the age-related decline in cognitive function in healthy middle-aged and elderly adults but the results also suggested that C. asiatica can modulate cognitive function differently at different time periods [115]. Recent pharmacokinetic studies with a standardised hot water extract of C. asiatica, when given to healthy older adults (white, Caucasian), demonstrated a significant difference in Cmax between male and female participants for the 2 g dose [5]. When a 60% ethanolic dry extract of Dandelion (Taraxacum officinale (L.) Weber) corresponding to 6.3 g leaf/day was tested in ten individuals (5F;5 M) for seven days and pooled for male and female participants, a more pronounced increase in the relative amount of Tamm-Horsfall protein/creatinine was observed on day three in women than men [116]. This could be possibly explained as a self‐defence mechanism to overcome the higher infections pressure by the female anatomical properties, since women have a tendency to have higher Tamm-Horsfall protein (THP; Uromodulin) concentration and excretion rates than men [117]. The effect of a Hypericum perforatum extract [300 mg containing 900 mg hypericin] taken three times a day for 14 days by healthy male and female participants, demonstrated an induction of the enzyme CYP1A2 only in females [118]. When Liquorice (Glycyrrhiza glabra L.) 100 g (containing 150 mg glycyrrhetinic acid) was consumed daily for four weeks, the relative change in serum aldosterone levels, showed men being more responsive than women, but not in patients with essential hypertension compared to healthy subjects [119]. A gender-specific benefit was also found with a standardized extract of ginger over placebo in Functional Living Index Emesis [FLIE] in female cancer patients receiving high-dose cisplatin [120].
Evaluation of the 150 human studies using ginger (pharmacological studies + clinical trials) showed that they were carried out in 25 countries. The main contributors were: Iran 39.6%, Thailand 8.7%, the US 8.1%, Europe collectively 17.4%, East Asia collectively 5.4% and the Indian subcontinent 5.4%. Gender analysis showed that (where gender was given) 55.5% of all studies included both men and women, 8% included men only, and 36.5% used only women. Trials for female-only conditions (NVP and primary dysmenorrhoea) represented 15.3%. Regarding the quality of ginger used in these studies, 53% were found to have used commercially available products.
Concluding Remarks
Ginger has a long tradition of benefit for various human ailments and although evidence has been scarce, recent meta-analyses and systematic reviews, together with the large number of clinical trials, now suggest effectiveness in [i] gastrointestinal disorders [121] and [ii] pain and metabolic syndrome [53], [54], [55], [122]. Similarly for nausea and vomiting in [iii] early pregnancy [123], [124], [125], [126], [iv] as an adjunct antiemetic in cancer chemotherapy [120], [127], [128], [129], [130] and [v] post-operatively [131], [132], [133]. However, as an antiemetic in the prevention of motion-induced sickness, observed results have been mixed depending on the experimental conditions used, i.e. travel or simulator [134], [135]. Promising results have however been reported for [vi] painful inflammatory conditions such as osteoarthritis [54], [55], [136], [137], [vii] neurodegenerative diseases [138], as well as for [viii] glycaemic control in type 2 diabetes mellitus and metabolic syndrome [139], [140], [141], [142] (see [Fig. 1]).


Many publications argue that such positive outcomes can be explained by the fact that ginger constituents interact with receptors of neurotransmitters mediating nausea and vomiting [143], as well as COX-2 associated with inflammation [144] and have anti-nociceptive activity involved in pain [122], [145], thereby proving activity. Similarly in silico mechanistic, in vitro and in vivo animal studies are indicative of supporting the findings from human clinical trials [53], [137]. However, such positive health benefits need to be interpreted with caution. Few clinical trials meet the criterion of ʼhigh qualityʼ, with too many small patient sizes and unstandardized evaluation systems including ginger quality and dosage regimes in the long-term [54], [55], [146]. Also, since most of the included trials were conducted in Asia, pharmacogenetics and outcome expectancy regarding ginger intervention could differ across cultures and ethnicities [137] and although there is an EMA guidance document concerning Ethnic Factors in the Acceptability of Foreign Clinical Data [147] this is rarely applied to herbal medicinal products. Finally, although ginger is a ʼuniversally usedʼ medicinal plant, surveys from around the world – Taiwan [148], Australia [149], Brazil [150], the US [151] and Europe [152] – show that women continue to be the primary users for conditions including stress, depression, menopause, pregnancy associated nausea, primary dysmenorrhoea and arthritis. Few references specifically relate to effects or recommended dosages for men, and such facts need to be reflected in the genders that are used for animal in vivo and human clinical trials.
Contributorsʼ Statement
Conception and Design of the work: I. Hook, L. Krenn, B. Steinhoff, E. Wolfram; Data collection, analysis and interpretation of the data and drafting the manuscript: I. Hook; Critical discussion and revision of the manuscript: L. Krenn, B. Steinhoff, E. Wolfram
Conflict of Interest
The authors declare that they have no conflict of interest.
Acknowledgements
The authors thank other members of the Scientific Committee of the European Scientific Cooperative on Phytotherapy for their input (ESCOP, https://escop.com/about-escop/scientific-committee/).
-
References
- 1 Kroes BH. The legal framework governing the quality of (traditional) herbal medicinal products in the European Union. J Ethnopharmacol 2014; 158 Pt B: 449-453
- 2 Parveen A, Parveen B, Parveen R, Ahmad S. Challenges and guidelines for clinical trial of herbal drugs. J Pharm Bioallied Sci 2015; 7: 329-333
- 3 Wang H, Chen Y, Wang L, Liu Q, Yang S, Wang C. Advancing herbal medicine: Enhancing product quality and safety through robust quality control practices. Front Pharmacol 2023; 14: 1265178
- 4 Zhang AL, Changli Xue C, Fong HHS. Ch 22 Integration of Herbal Medicine into Evidence-Based Clinical Practice: Current Status and Issues. In: Benzie IFF, Wachtel-Galor S. editors Herbal Medicine: Biomolecular and Clinical Aspects. 2nd ed.. Boca Raton (FL): CRC Press/Taylor & Francis; 2011
- 5 Wright KM, Bollen M, David J, Speers AB, Brandes MS, Gray NE, Alcázar Magaña A, McClure C, Stevens JF, Maier CS, Quinn JF, Soumyanath A. Pharmacokinetics and pharmacodynamics of key components of a standardized Centella asiatica product in cognitively impaired older adults: A phase 1, double-blind, randomized clinical trial. Antioxidants (Basel) 2022; 11: 215
- 6 Salm S, Rutz J, van den Akker M, Blaheta RA, Bachmeier BE. Current state of research on the clinical benefits of herbal medicines for non-life-threatening ailments. Front Pharmacol 2023; 14: 1234701
- 7 Huo H, Liu Y, Liu W, Sun J, Zhang Q, Zhao Y, Zheng J, Tu P, Song Y, Li J. A full solution for multi-component quantification-oriented quality assessment of herbal medicines, Chinese agarwood as a case. J Chromatogr A 2018; 1558: 37-49
- 8 Indrayanto G. The importance of method validation in herbal drug research. J Pharm Biomed Anal 2022; 214: 114735
- 9 Sharma S, Shukla MK, Sharma KC, Tirath, Kumar L, Anal JMH, Upadhyay SK, Bhattacharyya S, Kumar D. Revisiting the therapeutic potential of gingerols against different pharmacological activities. Naunyn Schmiedebergs Arch Pharmacol 2023; 396: 633-647
- 10 Mahboubi M. Zingiber officinale Rosc. essential oil, a review on its composition and bioactivity. Clin Phytosci 2019; 5: 6
- 11 Heinrich M. Quality and safety of herbal medical products: Regulation and the need for quality assurance along the value chains. Br J Clin Pharmacol 2015; 80: 62-66
- 12 The Pharmacopoeia of the Peopleʼs Republic of China, 2020. Accessed July 3, 2024 at: https://www.webofpharma.com/2023/09/chinese-pharmacopoeia-2020-in-english.html
- 13 European Pharmacopoeia. 7th ed. Ginger – Zingiberis rhizoma. Council of Europe 01/2011:1522 corrected 10.0
- 14 United States Pharmacopeia. Dietary Supplement Monographs, Ginger. USP-NF. Rockville, MD: United States Pharmacopeia; 2024
- 15 Bilia AR, Costa MDC. Medicinal plants and their preparations in the European market: Why has the harmonization failed? The cases of St. Johnʼs wort, valerian, ginkgo, ginseng, and green tea. Phytomedicine 2021; 81: 153421
- 16 Anon. Assessment Report on Zingiber officinale Roscoe, Rhizome. EMA/HMPC/885789/2022. Accessed July 3, 2024 at: https://www.ema.europa.eu/en/documents/herbal-report/draft-assessment-report-zingiber-officinale-roscoe-rhizoma-revision-1_en.pdf
- 17 Anon. The European market potential for dried ginger. Accessed January 26, 2025 at: https://www.cbi.eu/market-information/spices-herbs/dried-ginger/market-potential
- 18 Anon. Committee on Toxicity of Chemicals in Food, Consumer Products and the Environment. Accessed on September 2 2025 at: https://cot.food.gov.uk/sites/default/files/2021-05/TOX-2021-26%2520Safety%2520of%2520Ginger%2520Use%2520in%2520Pregnancy.pdf
- 19 Smith T, Carly L, Craft E. US Sales of Herbal Supplements increase by 4.4 % in 2023. Accessed March 17, 2025 at: https://umb.herbalgram.org/media/jk4f3rzm/hg141-hmr-web.pdf
- 20 Husain I, Zulfiqar A, Chittiboyina A, Khan I, Noonan G, Khan S. Modulation of PXR activity by ginger and its constituents. Accessed July 11, 2024 at: https://www.fda.gov/science-research/fda-science-forum/2021-fda-science-forum-05262021-05272021
- 21 Schwertner HA, Rios DC, Pascoe JE. Variation in concentration and labeling of ginger root dietary supplements. Obstet Gynecol 2006; 107: 1337-1343
- 22 Anon. Ginger Supplements, Chews & Spices Review. Accessed April 26, 2022 at: https://www.consumerlab.com/reviews/ginger-supplement-review/ginger/
- 23 Cui K, Wang J, Ma G, Guan S, Liang J, Fang L, Ding R, Li T, Dong Z, Wu X, Zheng Y. Residue levels, processing factors and risk assessment of pesticides in ginger from market to table. J Hazard Mater 2024; 470: 134268
- 24 Klier B, Knödler M, Peschke J, Riegert U, Steinhoff B. Pesticide residues in herbal drugs: Evaluation of a database. J Appl Res Med Aromat Plants 2019; 15: 100223
- 25 Luo L, Wang B, Jiang J, Fitzgerald M, Huang Q, Yu Z, Li H, Zhang J, Wei J, Yang C, Zhang H, Dong L, Chen S. Heavy metal contaminations in herbal medicines: Determination, comprehensive risk assessments, and solutions. Front Pharmacol 2021; 11: 595335
- 26 Bustami NA, Ho YB, Tan CK, Aris AZ, Tan ES. Heavy metals in danggui (Angelica sinensis) consumed by postpartum mothers and its health risk. Malaysian Journal of Medicine and Health Sciences 2020; 16: 113-117
- 27 Yin J, Zhuang J, Zhang X, Xu C, Lv S. Ginseng of different ages is affected by the accumulation of heavy metals in ginseng soil. PLoS One 2022; 17: e0269238
- 28 Giebułtowicz J, Ślusarczyk J, Wyderska S, Wroczyński P. The impact of organic selenium (IV) on Hypericum perforatum L. under cadmium stress and non-stress conditions. Plants (Basel) 2024; 13: 2099
- 29 Masarovičová E, Kráľová K, Kummerová M. Principles of classification of medicinal plants as hyperaccumulators or excluders. Acta Physiol Plant 2010; 32: 823-829
- 30 Xu J, Zhang J, Lv Y, Xu K, Lu S, Liu X, Yang Y. Effect of soil mercury pollution on ginger (Zingiber officinale Roscoe): Growth, product quality, health risks and silicon mitigation. Ecotoxicol Environ Saf 2020; 195: 110472
- 31 Albert H, Klier B, Knödler M, Steinhoff B. Findings on the heavy metal content in herbal drugs and essential oils – an update. Pharmeur Bio Sci Notes 2018; 2018: 62-111
- 32 Binder G, Knödler M, Klier B, Zang P, Albert H, Barofsky A, Steinhoff B. Findings on the heavy metal content in herbal drugs and essential oils – An updated database evaluation. JARMAP 2025; 44: 100609
- 33 Busia K. Perspectives on animal experimentation in herbal medicine research: Ethical dilemmas and scientific progress. J Herb Med 2024; 46: 100903
- 34 Lee M, Shin H, Park M, Kim A, Cha S, Lee H. Systems pharmacology approaches to herbal medicine research: A brief review. BMB Rep 2022; 55: 417-428
- 35 Borse SP, Singh DP, Nivsarkar M. Understanding the relevance of herb-drug interaction studies with special focus on interplays: A prerequisite for integrative medicine. Porto Biomed J 2019; 4: e15
- 36 Zucker I, Prendergast BJ. Sex differences in pharmacokinetics predict adverse drug reactions in women. Biol Sex Differ 2020; 11: 32
- 37 Nunamaker EA, Turner PV. Unmasking the adverse impacts of sex bias on science and research animal welfare. Animals (Basel) 2023; 13: 2792
- 38 Yagihashi S. Contribution of animal models to diabetes research: Its history, significance, and translation to humans. J Diabetes Investig 2023; 14: 1015-1037
- 39 Daniels Gatward LF, Kennard MR, Smith LIF, King AJF. The use of mice in diabetes research: The impact of physiological characteristics, choice of model and husbandry practices. Diabet Med 2021; 38: e14711
- 40 Ge JF, Qi CC, Qiao JP, Wang CW, Zhou NJ. Sex differences in ICR mice in the Morris water maze task. Physiol Res 2013; 62: 107-117
- 41 Tsao CH, Wu KY, Su NC, Edwards A, Huang GJ. The influence of sex difference on behavior and adult hippocampal neurogenesis in C57BL/6 mice. Sci Rep 2023; 13: 17297
- 42 Kennard MR, Nandi M, Chapple S, King AJ. The glucose tolerance test in mice: Sex, drugs and protocol. Diabetes Obes Metab 2022; 24: 2241-2252
- 43 Dekker MJHJ, de Vries ST, Versantvoort CHM, Drost-van Velze EGE, Bhatt M, van Meer PJK, Havinga IK, Gispen-de Wied CC, Mol PGM. Sex proportionality in pre-clinical and clinical trials: An evaluation of 22 marketing authorization application dossiers submitted to the european medicines agency. Front Med (Lausanne) 2021; 8: 643028
- 44 Marshall LJ, Bailey J, Cassotta M, Herrmann K, Pistollato F. Poor translatability of biomedical research using animals – A narrative review. Altern Lab Anim 2023; 51: 102-135
- 45 Spanagel R. Ten points to improve reproducibility and translation of animal research. Front Behav Neurosci 2022; 16: 869511
- 46 Radu BM, Anuta V, Stoian G. Gender-differences in mice hypericin plasma levels upon long-term Hypericum administration. Open Nat Prod J 2009; 2: 68-70
- 47 Valvassori SS, Borges C, Bavaresco DV, Varela RB, Resende WR, Peterle BR, Arent CO, Budni J, Quevedo J. Hypericum perforatum chronic treatment affects cognitive parameters and brain neurotrophic factor levels. Braz J Psychiatry 2018; 40: 367-375
- 48 Gray NE, Harris CJ, Quinn JF, Soumyanath A. Centella asiatica modulates antioxidant and mitochondrial pathways and improves cognitive function in mice. J Ethnopharmacol 2016; 180: 78-86
- 49 Matthews DG, Caruso M, Alcazar Magana A, Wright KM, Maier CS, Stevens JF, Gray NE, Quinn JF, Soumyanath A. Caffeoylquinic acids in Centella asiatica reverse cognitive deficits in male 5XFAD alzheimerʼs disease model mice. Nutrients 2020; 12: 3488
- 50 Speers AB, García-Jaramillo M, Feryn A, Matthews DG, Lichtenberg T, Caruso M, Wright KM, Quinn JF, Stevens JF, Maier CS, Soumyanath A, Gray NE. Centella asiatica alters metabolic pathways associated with alzheimerʼs disease in the 5 xFAD mouse model of β-amyloid accumulation. Front Pharmacol 2021; 12: 788312
- 51 Nusier MK, Bataineh HN, Daradkah HM. Adverse effects of rosemary (Rosmarinus officinalis L.) on reproductive function in adult male rats. Exp Biol Med (Maywood) 2007; 232: 809-813
- 52 Haksar A, Sharma A, Chawla R, Kumar R, Arora R, Singh S, Prasad J, Gupta M, Tripathi RP, Arora MP, Islam F, Sharma RK. Zingiber officinale exhibits behavioral radioprotection against radiation-induced CTA in a gender-specific manner. Pharmacol Biochem Behav 2006; 84: 179-188
- 53 Crichton M, Marshall S, Marx W, Isenring E, Lohning A. Therapeutic health effects of ginger (Zingiber officinale): updated narrative review exploring the mechanisms of action. Nutr Rev 2023; 81: 1213-1224
- 54 Anh NH, Kim SJ, Long NP, Min JE, Yoon YC, Lee EG, Kim M, Kim TJ, Yang YY, Son EY, Yoon SJ, Diem NC, Kim HM, Kwon SW. Ginger on human health: A comprehensive systematic review of 109 randomized controlled trials. Nutrients 2020; 12: 157
- 55 Crichton M, Davidson AR, Innerarity C, Marx W, Lohning A, Isenring E, Marshall S. Orally consumed ginger and human health: An umbrella review. Am J Clin Nutr 2022; 115: 1511-1527
- 56 Patel P, Gosai H, Patel A. Impact of Dietary Habits, Ethnicity, and Geographical Provenance in Shaping Human Gut Microbiome Diversity. In: Kothari V, Kumar P, Ray S. eds Probiotics, Prebiotics, Synbiotics, and Postbiotics. Singapore: Springer; 2023
- 57 Bennett G, Bardon LA, Gibney ER. A comparison of dietary patterns and factors influencing food choice among ethnic groups living in one locality: A systematic review. Nutrients 2022; 14: 941
- 58 Zurita T, Kazmierski KFM, Wong L, Faulkner M, Kuo S, Huszti H, Rao U. Racial/ethnic differences in dietary intake among a diverse sample of adolescents: An experimental study. Pediatr Obes 2021; 16: e12823
- 59 Brooks AW, Priya S, Blekhman R, Bordenstein SR. Gut microbiota diversity across ethnicities in the United States. PLoS Biol 2018; 16: e2006842
- 60 Jain A, Li XH, Chen WN. Similarities and differences in gut microbiome composition correlate with dietary patterns of Indian and Chinese adults. AMB Express 2018; 8: 104
- 61 Kim CC, Parkar SG, Gopal PK. Developing infant gut microflora and complementary nutrition. J Roy Soc New Zealand 2020; 50: 384-396
- 62 Dwiyanto J, Hussain MH, Reidpath D. et al. Ethnicity influences the gut microbiota of individuals sharing a geographical location: a cross-sectional study from a middle-income country. Sci Rep 2021; 11: 2618
- 63 Borrello K, Lim U, Park SY, Monroe KR, Maskarinec G, Boushey CJ, Wilkens LR, Randolph TW, Le Marchand L, Hullar MA, Lampe JW. Dietary intake mediates ethnic differences in gut microbial composition. Nutrients 2022; 14: 660
- 64 Mallott EK, Sitarik AR, Leve LD, Cioffi C, Camargo Jr CA, Hasegawa K, Bordenstein SR. Human microbiome variation associated with race and ethnicity emerges as early as 3 months of age. PLoS Biol 2023; 21: e3002230
- 65 Stearns JC, Zulyniak MA, de Souza RJ, Campbell NC, Fontes M, Shaikh M, Sears MR, Becker AB, Mandhane PJ, Subbarao P, Turvey SE, Gupta M, Beyene J, Surette MG, Anand SS. NutriGen Alliance. Ethnic and diet-related differences in the healthy infant microbiome. Genome Med 2017; 9: 32
- 66 Crichton M, Marshall S, Marx W, Isenring E, Vázquez-Campos X, Dawson SL, Lohning A. Effect of ginger root powder on gastrointestinal bacteria composition, gastrointestinal symptoms, mental health, fatigue, and quality of life: A double-blind placebo-controlled trial. J Nutr 2023; 153: 3193-3206
- 67 Prakash A, Rubin N, Staley C, Onyeaghala G, Wen YF, Shaukat A, Milne G, Straka RJ, Church TR, Prizment A. Effect of ginger supplementation on the fecal microbiome in subjects with prior colorectal adenoma. Sci Rep 2024; 14: 2988
- 68 Wang X, Zhang D, Jiang H, Zhang S, Pang X, Gao S, Zhang H, Zhang S, Xiao Q, Chen L, Wang S, Qi D, Li Y. Gut microbiota variation with short-term intake of ginger juice on human health. Front Microbiol 2021; 11: 576061
- 69 Johnson JA. Predictability of the effects of race or ethnicity on pharmacokinetics of drugs. Int J Clin Pharmacol Ther 2000; 38: 53-60
- 70 Alolga RN, Fan Y, Zhang G, Li J, Zhao YJ, Lelu Kakila J, Chen Y, Li P, Qi LW. Pharmacokinetics of a multicomponent herbal preparation in healthy Chinese and African volunteers. Sci Rep 2015; 5: 12961
- 71 Yu Y, Zick S, Li X, Zou P, Wright B, Sun D. Examination of the pharmacokinetics of active ingredients of ginger in humans. AAPS J 2011; 13: 417-426
- 72 Zick SM, Djuric Z, Ruffin MT, Litzinger AJ, Normolle DP, Alrawi S, Feng MR, Brenner DE. Pharmacokinetics of 6-gingerol, 8-gingerol, 10-gingerol, and 6-shogaol and conjugate metabolites in healthy human subjects. Cancer Epidemiol Biomarkers Prev 2008; 17: 1930-1936
- 73 Levita J, Syafitri DM, Supu RD, Mutakin M, Megantara S, Febrianti M, Diantini A. Pharmacokinetics of 10-gingerol and 6-shogaol in the plasma of healthy subjects treated with red ginger (Zingiber officinale var. Rubrum) suspension. Biomed Rep 2018; 9: 474-482
- 74 Rasoanaivo P, Wright CW, Willcox ML, Gilbert B. Whole plant extracts versus single compounds for the treatment of malaria: Synergy and positive interactions. Malar J 2011; 10: S4
- 75 Elfawal MA, Towler MJ, Reich NG, Weathers PJ, Rich SM. Dried whole-plant Artemisia annua slows evolution of malaria drug resistance and overcomes resistance to artemisinin. Proc Natl Acad Sci U S A 2015; 112: 821-826
- 76 Gundala SR, Mukkavilli R, Yang C, Yadav P, Tandon V, Vangala S, Prakash S, Aneja R. Enterohepatic recirculation of bioactive ginger phytochemicals is associated with enhanced tumor growth-inhibitory activity of ginger extract. Carcinogenesis 2014; 35: 1320-1329
- 77 Hengjumrut P, Anukunwithaya T, Tantisira MH, Tantisira B, Khemawoot P. Comparative pharmacokinetics between madecassoside and asiaticoside presented in a standardised extract of Centella asiatica, ECa 233 and their respective pure compound given separately in rats. Xenobiotica 2018; 48: 18-27
- 78 Anon. Inclusion of Women and Minorities as Participants in Research Involving Human Subjects. Accessed March 8, 2025 at: https://grants.nih.gov/policy-and-compliance/policy-topics/inclusion/women-and-minorities/guideline%23:~:text=If%2520the%2520data%2520from%2520prior%2520studies%2520neither%2520strongly%2520support%2520nor,clinical%2520trial%2520will%2520be%2520required
- 79 Chu T. Gender differences in pharmacokinetics. US Pharm 2014; 39: 40-43
- 80 Baggio G, Corsini A, Floreani A, Giannini S, Zagonel V. Gender medicine: A task for the third millennium. Clin Chem Lab Med 2013; 51: 713-727
- 81 Phillips SP, Hamberg K. Doubly blind: A systematic review of gender in randomised controlled trials. Glob Health Action 2016; 9: 29597
- 82 Franconi F, Campesi I, Colombo D, Antonini P. Sex-gender variable: Methodological recommendations for increasing scientific value of clinical studies. Cells 2019; 8: 476
- 83 Ravindran TS, Teerawattananon Y, Tannenbaum C, Vijayasingham L. Making pharmaceutical research and regulation work for women. BMJ 2020; 371: m3808
- 84 Soldin OP, Mattison DR. Sex differences in pharmacokinetics and pharmacodynamics. Clin Pharmacokinet 2009; 48: 143-157
- 85 Mauvais-Jarvis F, Bairey Merz N, Barnes PJ, Brinton RD, Carrero JJ, DeMeo DL, De Vries GJ, Epperson CN, Govindan R, Klein SL, Lonardo A, Maki PM, McCullough LD, Regitz-Zagrosek V, Regensteiner JG, Rubin JB, Sandberg K, Suzuki A. Sex and gender: Modifiers of health, disease, and medicine. Lancet 2020; 396: 565-582
- 86 Romanescu M, Buda V, Lombrea A, Andor M, Ledeti I, Suciu M, Danciu C, Dehelean CA, Dehelean L. Sex-related differences in pharmacological response to CNS drugs: A narrative review. J Pers Med 2022; 12: 907
- 87 Park AH, Hu S. Gender differences in motion sickness history and susceptibility to optokinetic rotation-induced motion sickness. Aviat Space Environ Med 1999; 70: 1077-1080
- 88 Schmidt EA, Kuiper OX, Wolter S, Diels C, Bos JE. An international survey on the incidence and modulating factors of carsickness. Transp Res Part F Traffic Psychol Behav 2020; 71: 76-87
- 89 Stern RM, Hu S, Uijtdehaage SH, Muth ER, Xu LH, Koch KL. Asian hypersusceptibility to motion sickness. Hum Hered 1996; 46: 7-14
- 90 Klosterhalfen S, Kellermann S, Pan F, Stockhorst U, Hall G, Enck P. Effects of ethnicity and gender on motion sickness susceptibility. Aviat Space Environ Med 2005; 76: 1051-1057
- 91 Chan A, Malhotra C, Do YK, Malhotra R, Ostbye T. Self-reported pain severity among multiethnic older Singaporeans: does adjusting for reporting heterogeneity matter?. Eur J Pain 2011; 15: 1094-1099
- 92 Campbell CM, Edwards RR. Ethnic differences in pain and pain management. Pain Manag 2012; 2: 219-230
- 93 Hesketh PJ, Grunberg SM, Herrstedt J, de Wit R, Gralla RJ, Carides AD, Taylor A, Evans JK, Horgan KJ. Combined data from two phase III trials of the NK1 antagonist aprepitant plus a 5 HT3 antagonist and a corticosteroid for prevention of chemotherapy-induced nausea and vomiting: effect of gender on treatment response. Support Care Cancer 2006; 14: 354
- 94 Salazar-Parra M, Guzman-Ramirez BG, Pintor-Belmontes KJ, Barbosa-Camacho FJ, Bernal-Hernández A, Cruz-Neri RU, Fuentes-Orozco C, Aguirre LLR, Rodríguez-Navarro D, Brancaccio-Pérez IV, Esparza-Estrada I, Cervantes-Pérez E, Cervantes-Guevara G, Cervantes-Cardona GA, González-Ojeda A. Gender differences in postoperative pain, nausea and vomiting after elective laparoscopic cholecystectomy. World J Surg 2020; 44: 4070-4076
- 95 Alli A, Omar S, Tsang S, Naik BI. The effect of ethnicity on the incidence of postoperative nausea and vomiting in moderate to high risk patients undergoing general anesthesia in South Africa: A controlled observational study. Middle East J Anaesthesiol 2017; 24: 119-129
- 96 Woodrow KM, Friedman GD, Siegelaub AB, Collen MF. Pain tolerance: Differences according to age, sex and race. Psychosom Med 1972; 34: 548-556
- 97 Bartley EJ, Fillingim RB. Sex differences in pain: A brief review of clinical and experimental findings. Br J Anaesth 2013; 111: 52-58
- 98 Taenzer AH, Clark C, Curry CS. Gender affects report of pain and function after arthroscopic anterior cruciate ligament reconstruction. Anesthesiology 2000; 93: 670-675
- 99 Pieretti S, Di Giannuario A, Di Giovannandrea R, Marzoli F, Piccaro G, Minosi P, Aloisi AM. Gender differences in pain and its relief. Ann Ist Super Sanita 2016; 52: 184-189
- 100 Rossi MF, Tumminello A, Marconi M, Gualano MR, Santoro PE, Malorni W, Moscato U. Sex and gender differences in migraines: A narrative review. Neurol Sci 2022; 43: 5729-5734
- 101 Segal NA, Nilges JM, Oo WM. Sex differences in osteoarthritis prevalence, pain perception, physical function and therapeutics. Osteoarthritis Cartilage 2024; 32: 1045-1053
- 102 Dabbous A, Itani M, Kawas N, Karam V, Aouad M, Baraka A, Khoury SJ, Khoury G. Post-laparoscopic vomiting in females versus males: Comparison of prophylactic antiemetic action of ondansetron versus metoclopramide. JSLS 1998; 2: 273-276
- 103 Brettner F, Janitza S, Prüll K, Weninger E, Mansmann U, Küchenhoff H, Jovanovic A, Pollwein B, Chappell D, Zwissler B, von Dossow V. Gender-specific differences in low-dose haloperidol response for prevention of postoperative nausea and vomiting: A register-based cohort study. PLoS One 2016; 11: e0146746
- 104 Mawhinney LJ, Mabourakh D, Lewis MC. Gender-specific differences in the central nervous systemʼs response to anesthesia. Transl Stroke Res 2013; 4: 462-475
- 105 Daoust R, Paquet J, Lavigne G, Piette E, Chauny JM. Impact of age, sex and route of administration on adverse events after opioid treatment in the emergency department: A retrospective study. Pain Res Manag 2015; 20: 23-28
- 106 Li M, Lu S, Wang G, Zhong N. The effects of gender differences in patients with depression on their emotional working memory and emotional experience. Behav Neurol 2015; 2015: 807343
- 107 Alcalde E, Rouquette A, Wiernik E, Rigal L. How do men and women differ in their depressive symptomatology? A gendered network analysis of depressive symptoms in a French population-based cohort. J Affect Disord 2024; 353: 1-10
- 108 Seeman MV. The pharmacodynamics of antipsychotic drugs in women and men. Front Psychiatry 2021; 12: 650904
- 109 Klein SL, Flanagan KL. Sex differences in immune responses. Nat Rev Immunol 2016; 16: 626-638
- 110 Wegner A, Benson S, Rebernik L, Spreitzer I, Jäger M, Schedlowski M, Elsenbruch S, Engler H. Sex differences in the pro-inflammatory cytokine response to endotoxin unfold in vivo but not ex vivo in healthy humans. Innate Immun 2017; 23: 432-439
- 111 Ingersoll MA. Sex differences shape the response to infectious diseases. PLoS Pathog 2017; 13: e1006688
- 112 Fitzgerald M, Culbert T, Finkelstein M, Green M, Johnson A, Chen S. The effect of gender and ethnicity on childrenʼs attitudes and preferences for essential oils: A pilot study. Explore (NY) 2007; 3: 378-385
- 113 Chen J, Zhang N, Pei S, Yao L. Odor perception of aromatherapy essential oils with different chemical types: Influence of gender and two cultural characteristics. Front Psychol 2022; 13: 998612
- 114 Campesi I, Romani A, Franconi F. The sex-gender effects in the road to tailored botanicals. Nutrients 2019; 11: 1637
- 115 Dev R, Dev O, Mohamed S, Hambali Z, Samah BA. Comparison on cognitive effects of Centella asiatica in healthy middle age female and male volunteers. Eur J Sci Res 2009; 31: 553-565
- 116 Mo B, Sendker J, Herrmann F, Nowak S, Hensel A. Aqueous extract from Equisetum arvense stimulates the secretion of Tamm-Horsfall protein in human urine after oral intake. Phytomedicine 2022; 104: 154302
- 117 Mo B, Scharf B, Gutheil C, Letzel MC, Hensel A. Tamm–Horsfall protein in humane urine: Sex-dependent differences in the excretion and N-glycosylation pattern. Sci Rep 2023; 13: 17815
- 118 Wenk M, Todesco L, Krähenbühl S. Effect of St Johnʼs wort on the activities of CYP1A2, CYP3A4, CYP2D6, N-acetyltransferase 2, and xanthine oxidase in healthy males and females. Br J Clin Pharmacol 2004; 57: 495-499
- 119 Sigurjonsdottir HA, Axelson M, Johannsson G, Manhem K, Nyström E, Wallerstedt S. The liquorice effect on the RAAS differs between the genders. Blood Press 2006; 15: 169-172
- 120 Bossi P, Cortinovis D, Fatigoni S, Cossu Rocca M, Fabi A, Seminara P, Ripamonti C, Alfieri S, Granata R, Bergamini C, Agustoni F, Bidoli P, Nolè F, Pessi MA, Macchi F, Michellini L, Montanaro F, Roila F. A randomized, double-blind, placebo-controlled, multicenter study of a ginger extract in the management of chemotherapy-induced nausea and vomiting (CINV) in patients receiving high-dose cisplatin. Ann Oncol 2017; 28: 2547-2551
- 121 Nikkhah Bodagh M, Maleki I, Hekmatdoost A. Ginger in gastrointestinal disorders: A systematic review of clinical trials. Food Sci Nutr 2018; 7: 96-108
- 122 Kim S, Cheon C, Kim B, Kim W. The effect of ginger and its sub-components on pain. Plants (Basel) 2022; 11: 2296
- 123 Ding M, Leach M, Bradley H. The effectiveness and safety of ginger for pregnancy-induced nausea and vomiting: A systematic review. Women Birth 2013; 26: e26-30
- 124 Thomson M, Corbin R, Leung L. Effects of ginger for nausea and vomiting in early pregnancy: A meta-analysis. J Am Board Fam Med 2014; 27: 115-122
- 125 Matthews A, Dowswell T, Haas DM, Doyle M, OʼMathúna DP. Interventions for nausea and vomiting in early pregnancy. Cochrane Database Syst Rev 2010; 9: CD007575. Update in: Cochrane Database Syst Rev 2014; (03) CD007575
- 126 Stanisiere J, Mousset PY, Lafay S. How safe is ginger rhizome for decreasing nausea and vomiting in women during early pregnancy?. Foods 2018; 7: 50
- 127 Lete I, Allué J. The effectiveness of ginger in the prevention of nausea and vomiting during pregnancy and chemotherapy. Integr Med Insights 2016; 11: 11-17
- 128 Chang WP, Peng YX. Does the oral administration of ginger reduce chemotherapy-induced nausea and vomiting? A meta-analysis of 10 randomized controlled trials. Cancer Nurs 2019; 42: E14-E23
- 129 Crichton M, Marshall S, Marx W, McCarthy AL, Isenring E. Efficacy of ginger (Zingiber officinale) in ameliorating chemotherapy-induced nausea and vomiting and chemotherapy-related outcomes: A systematic review update and meta-analysis. J Acad Nutr Diet 2019; 119: 2055-2068
- 130 Choi J, Lee J, Kim K, Choi HK, Lee SA, Lee HJ. Effects of ginger intake on chemotherapy-induced nausea and vomiting: A systematic review of randomized clinical trials. Nutrients 2022; 14: 4982
- 131 Zhu W, Dai Y, Huang M, Li J. Efficacy of ginger in preventing postoperative nausea and vomiting: A systematic review and meta-analysis. J Nurs Scholarsh 2021; 53: 671-679
- 132 Lu C, Chen X, Yan X, He J, Nie Z. The preventive and relieving effects of ginger on postoperative nausea and vomiting: A systematic review and meta-analysis of randomized controlled trials. Int J Nurs Stud 2022; 125: 104094
- 133 Zhao C, Chen W, Wang D, Cong X, Zhu M, Zhu C, Xu J, Cai J. Ginger (Zingiber officinale Roscoe) preparations for prophylaxis of postoperative nausea and vomiting: A Bayesian network meta-analysis. J Ethnopharmacol 2023; 317: 116791
- 134 Lien HC, Sun WM, Chen YH, Kim H, Hasler W, Owyang C. Effects of ginger on motion sickness and gastric slow-wave dysrhythmias induced by circular vection. Am J Physiol Gastrointest Liver Physiol 2003; 284: G481-G489
- 135 Wang JQ, Qi RR, Pan LL, Zhou W, Zhang LL, Cai YL. Motion sickness and resting energy expenditure in Chinese male adults. Aerosp Med Hum Perform 2016; 87: 1-7
- 136 Morvaridzadeh M, Fazelian S, Agah S, Khazdouz M, Rahimlou M, Agh F, Potter E, Heshmati S, Heshmati J. Effect of ginger (Zingiber officinale) on inflammatory markers: A systematic review and meta-analysis of randomized controlled trials. Cytokine 2020; 135: 155224
- 137 Rondanelli M, Fossari F, Vecchio V, Gasparri C, Peroni G, Spadaccini D, Riva A, Petrangolini G, Iannello G, Nichetti M, Infantino V, Perna S. Clinical trials on pain lowering effect of ginger: A narrative review. Phytother Res 2020; 34: 2843-2856
- 138 Arcusa R, Villaño D, Marhuenda J, Cano M, Cerdà B, Zafrilla P. Potential role of ginger (Zingiber officinale Roscoe) in the prevention of neurodegenerative diseases. Front Nutr 2022; 9: 809621
- 139 Zhu J, Chen H, Song Z, Wang X, Sun Z. Effects of ginger (Zingiber officinale Roscoe) on type 2 diabetes mellitus and components of the metabolic syndrome: A systematic review and meta-analysis of randomized controlled trials. Evid Based Complement Alternat Med 2018; 2018: 5692962
- 140 Hasani H, Arab A, Hadi A, Pourmasoumi M, Ghavami A, Miraghajani M. Does ginger supplementation lower blood pressure? A systematic review and meta-analysis of clinical trials. Phytother Res 2019; 33: 1639-1647
- 141 Huang FY, Deng T, Meng LX, Ma XL. Dietary ginger as a traditional therapy for blood sugar control in patients with type 2 diabetes mellitus: A systematic review and meta-analysis. Medicine (Baltimore) 2019; 98: e15054
- 142 Gumbarewicz E, Jarząb A, Stepulak A, Kukula-Koch W. Zingiber officinale Rosc. in the treatment of metabolic syndrome disorders – A review of in vivo studies. Int J Mol Sci 2022; 23: 15545
- 143 Mao QQ, Xu XY, Cao SY, Gan RY, Corke H, Beta T, Li HB. Bioactive compounds and bioactivities of ginger (Zingiber officinale Roscoe). Foods 2019; 8: 185
- 144 Bischoff-Kont I, Primke T, Niebergall LS, Zech T, Fürst R. Ginger constituent 6-shogaol inhibits inflammation- and angiogenesis-related cell functions in primary human endothelial cells. Front Pharmacol 2022; 13: 844767
- 145 Andrei C, Zanfirescu A, Niţulescu GM, Negreş S. Understanding the molecular mechanisms underlying the analgesic effect of ginger. Nutraceuticals 2022; 2: 384-403
- 146 Marx W, McKavanagh D, McCarthy AL, Bird R, Ried K, Chan A, Isenring L. The effect of ginger (Zingiber officinale) on platelet aggregation: A systematic literature review. PLoS One 2015; 10: e0141119 Erratum in: PLoS One 2015; 10: e0143675 Erratum in: PLoS One 2015; 10: e0143675
- 147 EMA. Guidance Document: ICH Topic E 5 (R1) Ethnic Factors in the Acceptability of Foreign Clinical Data. (1998). Accessed January 21, 2025 at: https://www.ema.europa.eu/en/documents/scientific-guideline/ich-e-5-r1-ethnic-factors-acceptability-foreign-clinical-data-step-5_en.pdf
- 148 Shih CC, Liao CC, Su YC, Tsai CC, Lin JG. Gender differences in traditional Chinese medicine use among adults in Taiwan. PLoS One 2012; 7: e32540
- 149 Burnett AJ, Livingstone KM, Woods JL, McNaughton SA. Dietary supplement use among Australian adults: Findings from the 2011–2012 national nutrition and physical activity survey. Nutrients 2017; 9: 1248
- 150 de Moraes Mello Boccolini P, Siqueira Boccolini C. Prevalence of complementary and alternative medicine (CAM) use in Brazil. BMC Complement Med Ther 2020; 20: 51
- 151 Rashrash M, Schommer JC, Brown LM. Prevalence and predictors of herbal medicine use among adults in the united states. J Patient Exp 2017; 4: 108-113
- 152 Krsnik S, Erjavec K. Factors influencing use of medicinal herbs. J Patient Exp 2024; 11: 23743735241241181
Correspondence
Publication History
Received: 27 March 2025
Accepted after revision: 04 August 2025
Article published online:
08 September 2025
© 2025. Thieme. All rights reserved.
Georg Thieme Verlag KG
Oswald-Hesse-Straße 50, 70469 Stuttgart, Germany
-
References
- 1 Kroes BH. The legal framework governing the quality of (traditional) herbal medicinal products in the European Union. J Ethnopharmacol 2014; 158 Pt B: 449-453
- 2 Parveen A, Parveen B, Parveen R, Ahmad S. Challenges and guidelines for clinical trial of herbal drugs. J Pharm Bioallied Sci 2015; 7: 329-333
- 3 Wang H, Chen Y, Wang L, Liu Q, Yang S, Wang C. Advancing herbal medicine: Enhancing product quality and safety through robust quality control practices. Front Pharmacol 2023; 14: 1265178
- 4 Zhang AL, Changli Xue C, Fong HHS. Ch 22 Integration of Herbal Medicine into Evidence-Based Clinical Practice: Current Status and Issues. In: Benzie IFF, Wachtel-Galor S. editors Herbal Medicine: Biomolecular and Clinical Aspects. 2nd ed.. Boca Raton (FL): CRC Press/Taylor & Francis; 2011
- 5 Wright KM, Bollen M, David J, Speers AB, Brandes MS, Gray NE, Alcázar Magaña A, McClure C, Stevens JF, Maier CS, Quinn JF, Soumyanath A. Pharmacokinetics and pharmacodynamics of key components of a standardized Centella asiatica product in cognitively impaired older adults: A phase 1, double-blind, randomized clinical trial. Antioxidants (Basel) 2022; 11: 215
- 6 Salm S, Rutz J, van den Akker M, Blaheta RA, Bachmeier BE. Current state of research on the clinical benefits of herbal medicines for non-life-threatening ailments. Front Pharmacol 2023; 14: 1234701
- 7 Huo H, Liu Y, Liu W, Sun J, Zhang Q, Zhao Y, Zheng J, Tu P, Song Y, Li J. A full solution for multi-component quantification-oriented quality assessment of herbal medicines, Chinese agarwood as a case. J Chromatogr A 2018; 1558: 37-49
- 8 Indrayanto G. The importance of method validation in herbal drug research. J Pharm Biomed Anal 2022; 214: 114735
- 9 Sharma S, Shukla MK, Sharma KC, Tirath, Kumar L, Anal JMH, Upadhyay SK, Bhattacharyya S, Kumar D. Revisiting the therapeutic potential of gingerols against different pharmacological activities. Naunyn Schmiedebergs Arch Pharmacol 2023; 396: 633-647
- 10 Mahboubi M. Zingiber officinale Rosc. essential oil, a review on its composition and bioactivity. Clin Phytosci 2019; 5: 6
- 11 Heinrich M. Quality and safety of herbal medical products: Regulation and the need for quality assurance along the value chains. Br J Clin Pharmacol 2015; 80: 62-66
- 12 The Pharmacopoeia of the Peopleʼs Republic of China, 2020. Accessed July 3, 2024 at: https://www.webofpharma.com/2023/09/chinese-pharmacopoeia-2020-in-english.html
- 13 European Pharmacopoeia. 7th ed. Ginger – Zingiberis rhizoma. Council of Europe 01/2011:1522 corrected 10.0
- 14 United States Pharmacopeia. Dietary Supplement Monographs, Ginger. USP-NF. Rockville, MD: United States Pharmacopeia; 2024
- 15 Bilia AR, Costa MDC. Medicinal plants and their preparations in the European market: Why has the harmonization failed? The cases of St. Johnʼs wort, valerian, ginkgo, ginseng, and green tea. Phytomedicine 2021; 81: 153421
- 16 Anon. Assessment Report on Zingiber officinale Roscoe, Rhizome. EMA/HMPC/885789/2022. Accessed July 3, 2024 at: https://www.ema.europa.eu/en/documents/herbal-report/draft-assessment-report-zingiber-officinale-roscoe-rhizoma-revision-1_en.pdf
- 17 Anon. The European market potential for dried ginger. Accessed January 26, 2025 at: https://www.cbi.eu/market-information/spices-herbs/dried-ginger/market-potential
- 18 Anon. Committee on Toxicity of Chemicals in Food, Consumer Products and the Environment. Accessed on September 2 2025 at: https://cot.food.gov.uk/sites/default/files/2021-05/TOX-2021-26%2520Safety%2520of%2520Ginger%2520Use%2520in%2520Pregnancy.pdf
- 19 Smith T, Carly L, Craft E. US Sales of Herbal Supplements increase by 4.4 % in 2023. Accessed March 17, 2025 at: https://umb.herbalgram.org/media/jk4f3rzm/hg141-hmr-web.pdf
- 20 Husain I, Zulfiqar A, Chittiboyina A, Khan I, Noonan G, Khan S. Modulation of PXR activity by ginger and its constituents. Accessed July 11, 2024 at: https://www.fda.gov/science-research/fda-science-forum/2021-fda-science-forum-05262021-05272021
- 21 Schwertner HA, Rios DC, Pascoe JE. Variation in concentration and labeling of ginger root dietary supplements. Obstet Gynecol 2006; 107: 1337-1343
- 22 Anon. Ginger Supplements, Chews & Spices Review. Accessed April 26, 2022 at: https://www.consumerlab.com/reviews/ginger-supplement-review/ginger/
- 23 Cui K, Wang J, Ma G, Guan S, Liang J, Fang L, Ding R, Li T, Dong Z, Wu X, Zheng Y. Residue levels, processing factors and risk assessment of pesticides in ginger from market to table. J Hazard Mater 2024; 470: 134268
- 24 Klier B, Knödler M, Peschke J, Riegert U, Steinhoff B. Pesticide residues in herbal drugs: Evaluation of a database. J Appl Res Med Aromat Plants 2019; 15: 100223
- 25 Luo L, Wang B, Jiang J, Fitzgerald M, Huang Q, Yu Z, Li H, Zhang J, Wei J, Yang C, Zhang H, Dong L, Chen S. Heavy metal contaminations in herbal medicines: Determination, comprehensive risk assessments, and solutions. Front Pharmacol 2021; 11: 595335
- 26 Bustami NA, Ho YB, Tan CK, Aris AZ, Tan ES. Heavy metals in danggui (Angelica sinensis) consumed by postpartum mothers and its health risk. Malaysian Journal of Medicine and Health Sciences 2020; 16: 113-117
- 27 Yin J, Zhuang J, Zhang X, Xu C, Lv S. Ginseng of different ages is affected by the accumulation of heavy metals in ginseng soil. PLoS One 2022; 17: e0269238
- 28 Giebułtowicz J, Ślusarczyk J, Wyderska S, Wroczyński P. The impact of organic selenium (IV) on Hypericum perforatum L. under cadmium stress and non-stress conditions. Plants (Basel) 2024; 13: 2099
- 29 Masarovičová E, Kráľová K, Kummerová M. Principles of classification of medicinal plants as hyperaccumulators or excluders. Acta Physiol Plant 2010; 32: 823-829
- 30 Xu J, Zhang J, Lv Y, Xu K, Lu S, Liu X, Yang Y. Effect of soil mercury pollution on ginger (Zingiber officinale Roscoe): Growth, product quality, health risks and silicon mitigation. Ecotoxicol Environ Saf 2020; 195: 110472
- 31 Albert H, Klier B, Knödler M, Steinhoff B. Findings on the heavy metal content in herbal drugs and essential oils – an update. Pharmeur Bio Sci Notes 2018; 2018: 62-111
- 32 Binder G, Knödler M, Klier B, Zang P, Albert H, Barofsky A, Steinhoff B. Findings on the heavy metal content in herbal drugs and essential oils – An updated database evaluation. JARMAP 2025; 44: 100609
- 33 Busia K. Perspectives on animal experimentation in herbal medicine research: Ethical dilemmas and scientific progress. J Herb Med 2024; 46: 100903
- 34 Lee M, Shin H, Park M, Kim A, Cha S, Lee H. Systems pharmacology approaches to herbal medicine research: A brief review. BMB Rep 2022; 55: 417-428
- 35 Borse SP, Singh DP, Nivsarkar M. Understanding the relevance of herb-drug interaction studies with special focus on interplays: A prerequisite for integrative medicine. Porto Biomed J 2019; 4: e15
- 36 Zucker I, Prendergast BJ. Sex differences in pharmacokinetics predict adverse drug reactions in women. Biol Sex Differ 2020; 11: 32
- 37 Nunamaker EA, Turner PV. Unmasking the adverse impacts of sex bias on science and research animal welfare. Animals (Basel) 2023; 13: 2792
- 38 Yagihashi S. Contribution of animal models to diabetes research: Its history, significance, and translation to humans. J Diabetes Investig 2023; 14: 1015-1037
- 39 Daniels Gatward LF, Kennard MR, Smith LIF, King AJF. The use of mice in diabetes research: The impact of physiological characteristics, choice of model and husbandry practices. Diabet Med 2021; 38: e14711
- 40 Ge JF, Qi CC, Qiao JP, Wang CW, Zhou NJ. Sex differences in ICR mice in the Morris water maze task. Physiol Res 2013; 62: 107-117
- 41 Tsao CH, Wu KY, Su NC, Edwards A, Huang GJ. The influence of sex difference on behavior and adult hippocampal neurogenesis in C57BL/6 mice. Sci Rep 2023; 13: 17297
- 42 Kennard MR, Nandi M, Chapple S, King AJ. The glucose tolerance test in mice: Sex, drugs and protocol. Diabetes Obes Metab 2022; 24: 2241-2252
- 43 Dekker MJHJ, de Vries ST, Versantvoort CHM, Drost-van Velze EGE, Bhatt M, van Meer PJK, Havinga IK, Gispen-de Wied CC, Mol PGM. Sex proportionality in pre-clinical and clinical trials: An evaluation of 22 marketing authorization application dossiers submitted to the european medicines agency. Front Med (Lausanne) 2021; 8: 643028
- 44 Marshall LJ, Bailey J, Cassotta M, Herrmann K, Pistollato F. Poor translatability of biomedical research using animals – A narrative review. Altern Lab Anim 2023; 51: 102-135
- 45 Spanagel R. Ten points to improve reproducibility and translation of animal research. Front Behav Neurosci 2022; 16: 869511
- 46 Radu BM, Anuta V, Stoian G. Gender-differences in mice hypericin plasma levels upon long-term Hypericum administration. Open Nat Prod J 2009; 2: 68-70
- 47 Valvassori SS, Borges C, Bavaresco DV, Varela RB, Resende WR, Peterle BR, Arent CO, Budni J, Quevedo J. Hypericum perforatum chronic treatment affects cognitive parameters and brain neurotrophic factor levels. Braz J Psychiatry 2018; 40: 367-375
- 48 Gray NE, Harris CJ, Quinn JF, Soumyanath A. Centella asiatica modulates antioxidant and mitochondrial pathways and improves cognitive function in mice. J Ethnopharmacol 2016; 180: 78-86
- 49 Matthews DG, Caruso M, Alcazar Magana A, Wright KM, Maier CS, Stevens JF, Gray NE, Quinn JF, Soumyanath A. Caffeoylquinic acids in Centella asiatica reverse cognitive deficits in male 5XFAD alzheimerʼs disease model mice. Nutrients 2020; 12: 3488
- 50 Speers AB, García-Jaramillo M, Feryn A, Matthews DG, Lichtenberg T, Caruso M, Wright KM, Quinn JF, Stevens JF, Maier CS, Soumyanath A, Gray NE. Centella asiatica alters metabolic pathways associated with alzheimerʼs disease in the 5 xFAD mouse model of β-amyloid accumulation. Front Pharmacol 2021; 12: 788312
- 51 Nusier MK, Bataineh HN, Daradkah HM. Adverse effects of rosemary (Rosmarinus officinalis L.) on reproductive function in adult male rats. Exp Biol Med (Maywood) 2007; 232: 809-813
- 52 Haksar A, Sharma A, Chawla R, Kumar R, Arora R, Singh S, Prasad J, Gupta M, Tripathi RP, Arora MP, Islam F, Sharma RK. Zingiber officinale exhibits behavioral radioprotection against radiation-induced CTA in a gender-specific manner. Pharmacol Biochem Behav 2006; 84: 179-188
- 53 Crichton M, Marshall S, Marx W, Isenring E, Lohning A. Therapeutic health effects of ginger (Zingiber officinale): updated narrative review exploring the mechanisms of action. Nutr Rev 2023; 81: 1213-1224
- 54 Anh NH, Kim SJ, Long NP, Min JE, Yoon YC, Lee EG, Kim M, Kim TJ, Yang YY, Son EY, Yoon SJ, Diem NC, Kim HM, Kwon SW. Ginger on human health: A comprehensive systematic review of 109 randomized controlled trials. Nutrients 2020; 12: 157
- 55 Crichton M, Davidson AR, Innerarity C, Marx W, Lohning A, Isenring E, Marshall S. Orally consumed ginger and human health: An umbrella review. Am J Clin Nutr 2022; 115: 1511-1527
- 56 Patel P, Gosai H, Patel A. Impact of Dietary Habits, Ethnicity, and Geographical Provenance in Shaping Human Gut Microbiome Diversity. In: Kothari V, Kumar P, Ray S. eds Probiotics, Prebiotics, Synbiotics, and Postbiotics. Singapore: Springer; 2023
- 57 Bennett G, Bardon LA, Gibney ER. A comparison of dietary patterns and factors influencing food choice among ethnic groups living in one locality: A systematic review. Nutrients 2022; 14: 941
- 58 Zurita T, Kazmierski KFM, Wong L, Faulkner M, Kuo S, Huszti H, Rao U. Racial/ethnic differences in dietary intake among a diverse sample of adolescents: An experimental study. Pediatr Obes 2021; 16: e12823
- 59 Brooks AW, Priya S, Blekhman R, Bordenstein SR. Gut microbiota diversity across ethnicities in the United States. PLoS Biol 2018; 16: e2006842
- 60 Jain A, Li XH, Chen WN. Similarities and differences in gut microbiome composition correlate with dietary patterns of Indian and Chinese adults. AMB Express 2018; 8: 104
- 61 Kim CC, Parkar SG, Gopal PK. Developing infant gut microflora and complementary nutrition. J Roy Soc New Zealand 2020; 50: 384-396
- 62 Dwiyanto J, Hussain MH, Reidpath D. et al. Ethnicity influences the gut microbiota of individuals sharing a geographical location: a cross-sectional study from a middle-income country. Sci Rep 2021; 11: 2618
- 63 Borrello K, Lim U, Park SY, Monroe KR, Maskarinec G, Boushey CJ, Wilkens LR, Randolph TW, Le Marchand L, Hullar MA, Lampe JW. Dietary intake mediates ethnic differences in gut microbial composition. Nutrients 2022; 14: 660
- 64 Mallott EK, Sitarik AR, Leve LD, Cioffi C, Camargo Jr CA, Hasegawa K, Bordenstein SR. Human microbiome variation associated with race and ethnicity emerges as early as 3 months of age. PLoS Biol 2023; 21: e3002230
- 65 Stearns JC, Zulyniak MA, de Souza RJ, Campbell NC, Fontes M, Shaikh M, Sears MR, Becker AB, Mandhane PJ, Subbarao P, Turvey SE, Gupta M, Beyene J, Surette MG, Anand SS. NutriGen Alliance. Ethnic and diet-related differences in the healthy infant microbiome. Genome Med 2017; 9: 32
- 66 Crichton M, Marshall S, Marx W, Isenring E, Vázquez-Campos X, Dawson SL, Lohning A. Effect of ginger root powder on gastrointestinal bacteria composition, gastrointestinal symptoms, mental health, fatigue, and quality of life: A double-blind placebo-controlled trial. J Nutr 2023; 153: 3193-3206
- 67 Prakash A, Rubin N, Staley C, Onyeaghala G, Wen YF, Shaukat A, Milne G, Straka RJ, Church TR, Prizment A. Effect of ginger supplementation on the fecal microbiome in subjects with prior colorectal adenoma. Sci Rep 2024; 14: 2988
- 68 Wang X, Zhang D, Jiang H, Zhang S, Pang X, Gao S, Zhang H, Zhang S, Xiao Q, Chen L, Wang S, Qi D, Li Y. Gut microbiota variation with short-term intake of ginger juice on human health. Front Microbiol 2021; 11: 576061
- 69 Johnson JA. Predictability of the effects of race or ethnicity on pharmacokinetics of drugs. Int J Clin Pharmacol Ther 2000; 38: 53-60
- 70 Alolga RN, Fan Y, Zhang G, Li J, Zhao YJ, Lelu Kakila J, Chen Y, Li P, Qi LW. Pharmacokinetics of a multicomponent herbal preparation in healthy Chinese and African volunteers. Sci Rep 2015; 5: 12961
- 71 Yu Y, Zick S, Li X, Zou P, Wright B, Sun D. Examination of the pharmacokinetics of active ingredients of ginger in humans. AAPS J 2011; 13: 417-426
- 72 Zick SM, Djuric Z, Ruffin MT, Litzinger AJ, Normolle DP, Alrawi S, Feng MR, Brenner DE. Pharmacokinetics of 6-gingerol, 8-gingerol, 10-gingerol, and 6-shogaol and conjugate metabolites in healthy human subjects. Cancer Epidemiol Biomarkers Prev 2008; 17: 1930-1936
- 73 Levita J, Syafitri DM, Supu RD, Mutakin M, Megantara S, Febrianti M, Diantini A. Pharmacokinetics of 10-gingerol and 6-shogaol in the plasma of healthy subjects treated with red ginger (Zingiber officinale var. Rubrum) suspension. Biomed Rep 2018; 9: 474-482
- 74 Rasoanaivo P, Wright CW, Willcox ML, Gilbert B. Whole plant extracts versus single compounds for the treatment of malaria: Synergy and positive interactions. Malar J 2011; 10: S4
- 75 Elfawal MA, Towler MJ, Reich NG, Weathers PJ, Rich SM. Dried whole-plant Artemisia annua slows evolution of malaria drug resistance and overcomes resistance to artemisinin. Proc Natl Acad Sci U S A 2015; 112: 821-826
- 76 Gundala SR, Mukkavilli R, Yang C, Yadav P, Tandon V, Vangala S, Prakash S, Aneja R. Enterohepatic recirculation of bioactive ginger phytochemicals is associated with enhanced tumor growth-inhibitory activity of ginger extract. Carcinogenesis 2014; 35: 1320-1329
- 77 Hengjumrut P, Anukunwithaya T, Tantisira MH, Tantisira B, Khemawoot P. Comparative pharmacokinetics between madecassoside and asiaticoside presented in a standardised extract of Centella asiatica, ECa 233 and their respective pure compound given separately in rats. Xenobiotica 2018; 48: 18-27
- 78 Anon. Inclusion of Women and Minorities as Participants in Research Involving Human Subjects. Accessed March 8, 2025 at: https://grants.nih.gov/policy-and-compliance/policy-topics/inclusion/women-and-minorities/guideline%23:~:text=If%2520the%2520data%2520from%2520prior%2520studies%2520neither%2520strongly%2520support%2520nor,clinical%2520trial%2520will%2520be%2520required
- 79 Chu T. Gender differences in pharmacokinetics. US Pharm 2014; 39: 40-43
- 80 Baggio G, Corsini A, Floreani A, Giannini S, Zagonel V. Gender medicine: A task for the third millennium. Clin Chem Lab Med 2013; 51: 713-727
- 81 Phillips SP, Hamberg K. Doubly blind: A systematic review of gender in randomised controlled trials. Glob Health Action 2016; 9: 29597
- 82 Franconi F, Campesi I, Colombo D, Antonini P. Sex-gender variable: Methodological recommendations for increasing scientific value of clinical studies. Cells 2019; 8: 476
- 83 Ravindran TS, Teerawattananon Y, Tannenbaum C, Vijayasingham L. Making pharmaceutical research and regulation work for women. BMJ 2020; 371: m3808
- 84 Soldin OP, Mattison DR. Sex differences in pharmacokinetics and pharmacodynamics. Clin Pharmacokinet 2009; 48: 143-157
- 85 Mauvais-Jarvis F, Bairey Merz N, Barnes PJ, Brinton RD, Carrero JJ, DeMeo DL, De Vries GJ, Epperson CN, Govindan R, Klein SL, Lonardo A, Maki PM, McCullough LD, Regitz-Zagrosek V, Regensteiner JG, Rubin JB, Sandberg K, Suzuki A. Sex and gender: Modifiers of health, disease, and medicine. Lancet 2020; 396: 565-582
- 86 Romanescu M, Buda V, Lombrea A, Andor M, Ledeti I, Suciu M, Danciu C, Dehelean CA, Dehelean L. Sex-related differences in pharmacological response to CNS drugs: A narrative review. J Pers Med 2022; 12: 907
- 87 Park AH, Hu S. Gender differences in motion sickness history and susceptibility to optokinetic rotation-induced motion sickness. Aviat Space Environ Med 1999; 70: 1077-1080
- 88 Schmidt EA, Kuiper OX, Wolter S, Diels C, Bos JE. An international survey on the incidence and modulating factors of carsickness. Transp Res Part F Traffic Psychol Behav 2020; 71: 76-87
- 89 Stern RM, Hu S, Uijtdehaage SH, Muth ER, Xu LH, Koch KL. Asian hypersusceptibility to motion sickness. Hum Hered 1996; 46: 7-14
- 90 Klosterhalfen S, Kellermann S, Pan F, Stockhorst U, Hall G, Enck P. Effects of ethnicity and gender on motion sickness susceptibility. Aviat Space Environ Med 2005; 76: 1051-1057
- 91 Chan A, Malhotra C, Do YK, Malhotra R, Ostbye T. Self-reported pain severity among multiethnic older Singaporeans: does adjusting for reporting heterogeneity matter?. Eur J Pain 2011; 15: 1094-1099
- 92 Campbell CM, Edwards RR. Ethnic differences in pain and pain management. Pain Manag 2012; 2: 219-230
- 93 Hesketh PJ, Grunberg SM, Herrstedt J, de Wit R, Gralla RJ, Carides AD, Taylor A, Evans JK, Horgan KJ. Combined data from two phase III trials of the NK1 antagonist aprepitant plus a 5 HT3 antagonist and a corticosteroid for prevention of chemotherapy-induced nausea and vomiting: effect of gender on treatment response. Support Care Cancer 2006; 14: 354
- 94 Salazar-Parra M, Guzman-Ramirez BG, Pintor-Belmontes KJ, Barbosa-Camacho FJ, Bernal-Hernández A, Cruz-Neri RU, Fuentes-Orozco C, Aguirre LLR, Rodríguez-Navarro D, Brancaccio-Pérez IV, Esparza-Estrada I, Cervantes-Pérez E, Cervantes-Guevara G, Cervantes-Cardona GA, González-Ojeda A. Gender differences in postoperative pain, nausea and vomiting after elective laparoscopic cholecystectomy. World J Surg 2020; 44: 4070-4076
- 95 Alli A, Omar S, Tsang S, Naik BI. The effect of ethnicity on the incidence of postoperative nausea and vomiting in moderate to high risk patients undergoing general anesthesia in South Africa: A controlled observational study. Middle East J Anaesthesiol 2017; 24: 119-129
- 96 Woodrow KM, Friedman GD, Siegelaub AB, Collen MF. Pain tolerance: Differences according to age, sex and race. Psychosom Med 1972; 34: 548-556
- 97 Bartley EJ, Fillingim RB. Sex differences in pain: A brief review of clinical and experimental findings. Br J Anaesth 2013; 111: 52-58
- 98 Taenzer AH, Clark C, Curry CS. Gender affects report of pain and function after arthroscopic anterior cruciate ligament reconstruction. Anesthesiology 2000; 93: 670-675
- 99 Pieretti S, Di Giannuario A, Di Giovannandrea R, Marzoli F, Piccaro G, Minosi P, Aloisi AM. Gender differences in pain and its relief. Ann Ist Super Sanita 2016; 52: 184-189
- 100 Rossi MF, Tumminello A, Marconi M, Gualano MR, Santoro PE, Malorni W, Moscato U. Sex and gender differences in migraines: A narrative review. Neurol Sci 2022; 43: 5729-5734
- 101 Segal NA, Nilges JM, Oo WM. Sex differences in osteoarthritis prevalence, pain perception, physical function and therapeutics. Osteoarthritis Cartilage 2024; 32: 1045-1053
- 102 Dabbous A, Itani M, Kawas N, Karam V, Aouad M, Baraka A, Khoury SJ, Khoury G. Post-laparoscopic vomiting in females versus males: Comparison of prophylactic antiemetic action of ondansetron versus metoclopramide. JSLS 1998; 2: 273-276
- 103 Brettner F, Janitza S, Prüll K, Weninger E, Mansmann U, Küchenhoff H, Jovanovic A, Pollwein B, Chappell D, Zwissler B, von Dossow V. Gender-specific differences in low-dose haloperidol response for prevention of postoperative nausea and vomiting: A register-based cohort study. PLoS One 2016; 11: e0146746
- 104 Mawhinney LJ, Mabourakh D, Lewis MC. Gender-specific differences in the central nervous systemʼs response to anesthesia. Transl Stroke Res 2013; 4: 462-475
- 105 Daoust R, Paquet J, Lavigne G, Piette E, Chauny JM. Impact of age, sex and route of administration on adverse events after opioid treatment in the emergency department: A retrospective study. Pain Res Manag 2015; 20: 23-28
- 106 Li M, Lu S, Wang G, Zhong N. The effects of gender differences in patients with depression on their emotional working memory and emotional experience. Behav Neurol 2015; 2015: 807343
- 107 Alcalde E, Rouquette A, Wiernik E, Rigal L. How do men and women differ in their depressive symptomatology? A gendered network analysis of depressive symptoms in a French population-based cohort. J Affect Disord 2024; 353: 1-10
- 108 Seeman MV. The pharmacodynamics of antipsychotic drugs in women and men. Front Psychiatry 2021; 12: 650904
- 109 Klein SL, Flanagan KL. Sex differences in immune responses. Nat Rev Immunol 2016; 16: 626-638
- 110 Wegner A, Benson S, Rebernik L, Spreitzer I, Jäger M, Schedlowski M, Elsenbruch S, Engler H. Sex differences in the pro-inflammatory cytokine response to endotoxin unfold in vivo but not ex vivo in healthy humans. Innate Immun 2017; 23: 432-439
- 111 Ingersoll MA. Sex differences shape the response to infectious diseases. PLoS Pathog 2017; 13: e1006688
- 112 Fitzgerald M, Culbert T, Finkelstein M, Green M, Johnson A, Chen S. The effect of gender and ethnicity on childrenʼs attitudes and preferences for essential oils: A pilot study. Explore (NY) 2007; 3: 378-385
- 113 Chen J, Zhang N, Pei S, Yao L. Odor perception of aromatherapy essential oils with different chemical types: Influence of gender and two cultural characteristics. Front Psychol 2022; 13: 998612
- 114 Campesi I, Romani A, Franconi F. The sex-gender effects in the road to tailored botanicals. Nutrients 2019; 11: 1637
- 115 Dev R, Dev O, Mohamed S, Hambali Z, Samah BA. Comparison on cognitive effects of Centella asiatica in healthy middle age female and male volunteers. Eur J Sci Res 2009; 31: 553-565
- 116 Mo B, Sendker J, Herrmann F, Nowak S, Hensel A. Aqueous extract from Equisetum arvense stimulates the secretion of Tamm-Horsfall protein in human urine after oral intake. Phytomedicine 2022; 104: 154302
- 117 Mo B, Scharf B, Gutheil C, Letzel MC, Hensel A. Tamm–Horsfall protein in humane urine: Sex-dependent differences in the excretion and N-glycosylation pattern. Sci Rep 2023; 13: 17815
- 118 Wenk M, Todesco L, Krähenbühl S. Effect of St Johnʼs wort on the activities of CYP1A2, CYP3A4, CYP2D6, N-acetyltransferase 2, and xanthine oxidase in healthy males and females. Br J Clin Pharmacol 2004; 57: 495-499
- 119 Sigurjonsdottir HA, Axelson M, Johannsson G, Manhem K, Nyström E, Wallerstedt S. The liquorice effect on the RAAS differs between the genders. Blood Press 2006; 15: 169-172
- 120 Bossi P, Cortinovis D, Fatigoni S, Cossu Rocca M, Fabi A, Seminara P, Ripamonti C, Alfieri S, Granata R, Bergamini C, Agustoni F, Bidoli P, Nolè F, Pessi MA, Macchi F, Michellini L, Montanaro F, Roila F. A randomized, double-blind, placebo-controlled, multicenter study of a ginger extract in the management of chemotherapy-induced nausea and vomiting (CINV) in patients receiving high-dose cisplatin. Ann Oncol 2017; 28: 2547-2551
- 121 Nikkhah Bodagh M, Maleki I, Hekmatdoost A. Ginger in gastrointestinal disorders: A systematic review of clinical trials. Food Sci Nutr 2018; 7: 96-108
- 122 Kim S, Cheon C, Kim B, Kim W. The effect of ginger and its sub-components on pain. Plants (Basel) 2022; 11: 2296
- 123 Ding M, Leach M, Bradley H. The effectiveness and safety of ginger for pregnancy-induced nausea and vomiting: A systematic review. Women Birth 2013; 26: e26-30
- 124 Thomson M, Corbin R, Leung L. Effects of ginger for nausea and vomiting in early pregnancy: A meta-analysis. J Am Board Fam Med 2014; 27: 115-122
- 125 Matthews A, Dowswell T, Haas DM, Doyle M, OʼMathúna DP. Interventions for nausea and vomiting in early pregnancy. Cochrane Database Syst Rev 2010; 9: CD007575. Update in: Cochrane Database Syst Rev 2014; (03) CD007575
- 126 Stanisiere J, Mousset PY, Lafay S. How safe is ginger rhizome for decreasing nausea and vomiting in women during early pregnancy?. Foods 2018; 7: 50
- 127 Lete I, Allué J. The effectiveness of ginger in the prevention of nausea and vomiting during pregnancy and chemotherapy. Integr Med Insights 2016; 11: 11-17
- 128 Chang WP, Peng YX. Does the oral administration of ginger reduce chemotherapy-induced nausea and vomiting? A meta-analysis of 10 randomized controlled trials. Cancer Nurs 2019; 42: E14-E23
- 129 Crichton M, Marshall S, Marx W, McCarthy AL, Isenring E. Efficacy of ginger (Zingiber officinale) in ameliorating chemotherapy-induced nausea and vomiting and chemotherapy-related outcomes: A systematic review update and meta-analysis. J Acad Nutr Diet 2019; 119: 2055-2068
- 130 Choi J, Lee J, Kim K, Choi HK, Lee SA, Lee HJ. Effects of ginger intake on chemotherapy-induced nausea and vomiting: A systematic review of randomized clinical trials. Nutrients 2022; 14: 4982
- 131 Zhu W, Dai Y, Huang M, Li J. Efficacy of ginger in preventing postoperative nausea and vomiting: A systematic review and meta-analysis. J Nurs Scholarsh 2021; 53: 671-679
- 132 Lu C, Chen X, Yan X, He J, Nie Z. The preventive and relieving effects of ginger on postoperative nausea and vomiting: A systematic review and meta-analysis of randomized controlled trials. Int J Nurs Stud 2022; 125: 104094
- 133 Zhao C, Chen W, Wang D, Cong X, Zhu M, Zhu C, Xu J, Cai J. Ginger (Zingiber officinale Roscoe) preparations for prophylaxis of postoperative nausea and vomiting: A Bayesian network meta-analysis. J Ethnopharmacol 2023; 317: 116791
- 134 Lien HC, Sun WM, Chen YH, Kim H, Hasler W, Owyang C. Effects of ginger on motion sickness and gastric slow-wave dysrhythmias induced by circular vection. Am J Physiol Gastrointest Liver Physiol 2003; 284: G481-G489
- 135 Wang JQ, Qi RR, Pan LL, Zhou W, Zhang LL, Cai YL. Motion sickness and resting energy expenditure in Chinese male adults. Aerosp Med Hum Perform 2016; 87: 1-7
- 136 Morvaridzadeh M, Fazelian S, Agah S, Khazdouz M, Rahimlou M, Agh F, Potter E, Heshmati S, Heshmati J. Effect of ginger (Zingiber officinale) on inflammatory markers: A systematic review and meta-analysis of randomized controlled trials. Cytokine 2020; 135: 155224
- 137 Rondanelli M, Fossari F, Vecchio V, Gasparri C, Peroni G, Spadaccini D, Riva A, Petrangolini G, Iannello G, Nichetti M, Infantino V, Perna S. Clinical trials on pain lowering effect of ginger: A narrative review. Phytother Res 2020; 34: 2843-2856
- 138 Arcusa R, Villaño D, Marhuenda J, Cano M, Cerdà B, Zafrilla P. Potential role of ginger (Zingiber officinale Roscoe) in the prevention of neurodegenerative diseases. Front Nutr 2022; 9: 809621
- 139 Zhu J, Chen H, Song Z, Wang X, Sun Z. Effects of ginger (Zingiber officinale Roscoe) on type 2 diabetes mellitus and components of the metabolic syndrome: A systematic review and meta-analysis of randomized controlled trials. Evid Based Complement Alternat Med 2018; 2018: 5692962
- 140 Hasani H, Arab A, Hadi A, Pourmasoumi M, Ghavami A, Miraghajani M. Does ginger supplementation lower blood pressure? A systematic review and meta-analysis of clinical trials. Phytother Res 2019; 33: 1639-1647
- 141 Huang FY, Deng T, Meng LX, Ma XL. Dietary ginger as a traditional therapy for blood sugar control in patients with type 2 diabetes mellitus: A systematic review and meta-analysis. Medicine (Baltimore) 2019; 98: e15054
- 142 Gumbarewicz E, Jarząb A, Stepulak A, Kukula-Koch W. Zingiber officinale Rosc. in the treatment of metabolic syndrome disorders – A review of in vivo studies. Int J Mol Sci 2022; 23: 15545
- 143 Mao QQ, Xu XY, Cao SY, Gan RY, Corke H, Beta T, Li HB. Bioactive compounds and bioactivities of ginger (Zingiber officinale Roscoe). Foods 2019; 8: 185
- 144 Bischoff-Kont I, Primke T, Niebergall LS, Zech T, Fürst R. Ginger constituent 6-shogaol inhibits inflammation- and angiogenesis-related cell functions in primary human endothelial cells. Front Pharmacol 2022; 13: 844767
- 145 Andrei C, Zanfirescu A, Niţulescu GM, Negreş S. Understanding the molecular mechanisms underlying the analgesic effect of ginger. Nutraceuticals 2022; 2: 384-403
- 146 Marx W, McKavanagh D, McCarthy AL, Bird R, Ried K, Chan A, Isenring L. The effect of ginger (Zingiber officinale) on platelet aggregation: A systematic literature review. PLoS One 2015; 10: e0141119 Erratum in: PLoS One 2015; 10: e0143675 Erratum in: PLoS One 2015; 10: e0143675
- 147 EMA. Guidance Document: ICH Topic E 5 (R1) Ethnic Factors in the Acceptability of Foreign Clinical Data. (1998). Accessed January 21, 2025 at: https://www.ema.europa.eu/en/documents/scientific-guideline/ich-e-5-r1-ethnic-factors-acceptability-foreign-clinical-data-step-5_en.pdf
- 148 Shih CC, Liao CC, Su YC, Tsai CC, Lin JG. Gender differences in traditional Chinese medicine use among adults in Taiwan. PLoS One 2012; 7: e32540
- 149 Burnett AJ, Livingstone KM, Woods JL, McNaughton SA. Dietary supplement use among Australian adults: Findings from the 2011–2012 national nutrition and physical activity survey. Nutrients 2017; 9: 1248
- 150 de Moraes Mello Boccolini P, Siqueira Boccolini C. Prevalence of complementary and alternative medicine (CAM) use in Brazil. BMC Complement Med Ther 2020; 20: 51
- 151 Rashrash M, Schommer JC, Brown LM. Prevalence and predictors of herbal medicine use among adults in the united states. J Patient Exp 2017; 4: 108-113
- 152 Krsnik S, Erjavec K. Factors influencing use of medicinal herbs. J Patient Exp 2024; 11: 23743735241241181


