Isolation and analysis of ginseng: advances and challenges

Lian-Wen Qi *ab, Chong-Zhi Wang a and Chun-Su Yuan *a
aTang Center for Herbal Medicine Research and Department of Anesthesia & Critical Care, The Pritzker School of Medicine, The University of Chicago, 5841 South Maryland Avenue, Chicago, Illinois 60637, USA. E-mail: cyuan@dacc.uchicago.edu
bKey Laboratory of Modern Chinese Medicines (China Pharmaceutical University), Ministry of Education, Nanjing, 210009, China. E-mail: fleude@126.com

Received 15th October 2010

First published on 24th January 2011


Abstract

Covering: January 2000 to September 2010

Ginseng occupies a prominent position in the list of best-selling natural products in the world. Because of its complex constituents, multidisciplinary techniques are needed to validate the analytical methods that support ginseng's use worldwide. In the past decade, rapid development of technology has advanced many aspects of ginseng research. The aim of this review is to illustrate the recent advances in the isolation and analysis of ginseng, and to highlight new applications and challenges. Emphasis is placed on recent trends and emerging techniques.


Lian-Wen Qi

Lian-Wen Qi

Lian-Wen Qi is a research scholar in the Tang Center for Herbal Medicine Research at the University of Chicago. He obtained his Ph.D. in Pharmacognosy at China Pharmaceutical University. Since 2009, using multi-disciplinary approaches, he has been investigating plant fingerprinting and endogenous metabonomics for the quality control of different natural products, and characterizing the response of biological systems to a number of botanical components. Dr Qi's research interests include new technology development in phytochemistry, discovery of novel bioactive lead components, and their potential clinical applications. He has published over 50 peer-reviewed journal research papers, reviews and book chapters.

Chong-Zhi Wang

Chong-Zhi Wang

Chong-Zhi Wang is a Research Assistant Professor in the Department of Anesthesia and Critical Care, and serves as Associate Director of the Tang Center for Herbal Medicine Research and as Botanical Core Co-Leader of a NIH/NCCAM center project at the University of Chicago. He obtained his Ph.D. from China Pharmaceutical University and finished his postdoctoral training at the University of Chicago. Dr Wang's current work, in part supported by his NIH K01 grant, focuses on the identification of active botanical constituents and the evaluation of their pharmacological activities. He has published 73 peer-reviewed journal research articles, reviews and book chapters.

Chun-Su Yuan

Chun-Su Yuan

Chun-Su Yuan is the Cyrus Tang Professor in the Department of Anesthesia & Critical Care, and Director of the Tang Center for Herbal Medicine Research, University of Chicago. Since 1994, he has conducted clinical trials in evaluating the safety and efficacy of a novel compound, methylnaltrexone, approved in 2008 for opioid-induced constipation in advanced-illness patients. Dr Yuan also has a deep interest in botanical research, and has published over 200 articles. In addition, he currently serves as the Editor-in-Chief of the American Journal of Chinese Medicine.


1 Introduction

Plants have been the basis of traditional medicines for thousands of years, and continue to be considered valuable materials in medicines. Natural products isolated from plants provide an unparalleled source of chemical diversity for discovery of biologically active molecules. For various medical conditions, especially chronic diseases, use of medicinal plants has expanded globally and gained considerable popularity.1 Among these plants, ginseng has a long history and is today one of the world's most widely used medicinal plants. Ginseng products are commercially available in roots, tablets and capsules, liquid extracts, carbonated drinks and teas.2

The name ginseng is derived from a Chinese term referring to the “man-like” shape of the root. The genus is Panax, meaning “cure all” in Greek. Panax ginseng C. A. Meyer (commonly referred to as Asian, Chinese or Korean ginseng) and Panax quinquefolius L. (also known as American or North American ginseng) are the two most recognized species around the world. Another is Panax notoginseng (Burk.) F. H. Chen. Other less known species in the ginseng genus include Panax japonicus C.A. Meyer (Japanese ginseng) and Panax vietnamensis Ha et Grushv. (Vietnamese ginseng). Traditionally the ginseng root, available in white or red form, is used. White ginseng is prepared by air-drying after harvest, and red ginseng is prepared by a steaming or heating process.3,4 The leaf, berry and other parts of ginseng are also medicinal sources.5–7

Ginseng contains complex constituents. Thus, multidisciplinary methods are required to validate the analytical methods that support ginseng's use worldwide. In the past decade, advanced technologies for isolation and analysis of plants have revolutionized ginseng research. The literature describes investigation of ginseng with pharmacognosy, phytochemistry, biosynthesis, and pharmacology.8–13 The aim of this review is to illustrate the recent advances in the isolation and analysis of ginseng, and to highlight their new applications, especially in species authentication, quality assessment, and interaction with biological systems. This review covers the literature between January 2000 and September 2010, and is organized into three major sections: (1) preparation and isolation; (2) analytical advances; and (3) applications and challenges. Recent trends and emerging techniques are emphasized rather than routine methods. Information related to biosynthesis, pharmacology and clinical aspects is beyond the scope of this article.

2 Preparation and isolation

The first challenge in isolating and analyzing ginseng is the complexity of the sample matrices. The construction and maintenance of high-quality chemical libraries in ginseng is needed, calling for state-of-the-art procedures for separation and isolation. Recent developments in this field include automated and solvent-free extraction techniques and more efficient isolation methods.

2.1 Sample extraction and preparation techniques

Efficient sample preparation can improve extraction efficiency and enrich the target analytes. Ginseng saponins have been extracted from ginseng with different solvents and methods. The conventional method uses heat-reflux,14 Soxhlet,15 shaking16 or ultrasound-assisted extraction (UAE).17 Soxhlet extraction at 80–90 °C for 20–24 h efficiently extracts saponins.15 Modern ultra-pressure or ultra-temperature extraction methods have been applied such as pressurized liquid extraction (PLE),18 microwave-assisted extraction (MAE),19 high-pressure MAE,20 pressurized hot water extraction (PHWE)21 and supercritical fluid extraction (SFE).15 Compared with conventional methods, newer methods use less solvent, are easily automated, take a short time and are more efficient.22Table 1 lists the characteristics of nine different extraction techniques. Each has advantages and limitations depending on the projected use of results.
Table 1 Characteristics of nine extraction techniques for saponins from ginseng plant materials
Techniquea Core technology Extraction efficiency Operation Speed Automation Cost Ref.
a Abbreviations: HRE: heat reflux extraction; SAE: shaking-assisted extraction; UAE: ultrasound-assisted extraction; PLE: pressurized liquid extraction; MAE: microwave-assisted extraction; HP-MAE: high-pressure MAE; PHWE: pressurized hot- water extraction; SFE: supercritical fluid extraction.
HRE Heating Low Simple Low Difficult Very low 14
Soxhlet Soxhlet extractor High Moderate Very low Possible Low 15
SAE Mechanical shaking High Simple Moderate Difficult Low 16
UAE Ultrasound Low Simple Low Difficult Low 17
PLE High pressure Moderate Complicated Rapid Easy Moderate 18
MAE Microwave Moderate Simple Rapid Difficult Low 19
HP-MAE High pressure and microwave Moderate Simple Rapid Difficult Low 20
PHWE High pressure and hot water Moderate Complicated Moderate Easy Moderate 21
SFE Supercritical fluid Moderate Complicated Rapid Easy High 15


Plant material is considered first. Small sample particles have larger specific external areas, stronger solvent penetration, and thus higher extraction yield.23 Extraction solvent and extraction volume are key in obtaining high recovery.24 Among various solvent systems, 100% methanol has better extraction efficiency than water or 70% aqueous methanol extraction;25 50% ethanol has higher efficiency than an ethanolglycerinwater mixture or 65% glycerin.26 Adding non-ionic surfactants like Triton X-100 to solvent enhances the extraction yield.27 Various extraction methods have been compared quantitatively.18,23,28 Extraction conditions affect results, and the best conditions for each technique should be optimized in comparison. For instance, efficiency differs between Soxhlet extraction for 2 h23 and 20 h.15 Extraction efficiency also depends on the ginsenoside. Since degradation is possible during the extraction process, temperature is a factor to be considered.29

2.2 Isolation and purification methods

A high-quality chemical library of ginseng saponins is important for high-throughput screening, research into the structure–activity relationship, and investigation of the mechanisms of this group of compounds. Because ginseng has many constituents, the isolation process can be time-consuming and tedious. New technologies are therefore welcome.
2.2.1 Liquid–solid isolation. Separation and isolation of ginseng saponins is commonly performed with liquid–solid column chromatography.30,31Fig. 1 summarizes the procedures for isolation of saponins from ginseng materials. Ginseng samples are extracted with methanol30,31 or ethanol32,33 once or several times, and the combined extracts are concentrated in vacuo to dryness. The residue is suspended in water and then partitioned with organic solvents into different solvent-soluble portions such as n-hexane,30 dichloromethane,30,34 ethyl acetate,35n-butanol,36 and aqueous layers.31,34 The n-hexane and dichloromethane layers containing macromolecule and oil-soluble impurities are removed.30,34 The other portions are subjected to Diaion HP-20 column or silica gel column chromatography and eluted with a gradient solvent system to fractions and sub-fractions.31,34 The fractions are then repeatedly chromatographed with normal-phase silica gel column chromatography,31,35 reversed-phase silica gel column chromatography,31 and MCI-gel CHP20P columns,33 using a stepwise gradient elution of different solvent systems. The products are purified by preparative liquid chromatography,32,36 and their structures are identified by chemical and spectroscopic methods.
Procedures for isolation of ginseng saponins from ginseng plant materials.
Fig. 1 Procedures for isolation of ginseng saponins from ginseng plant materials.
2.2.2 Liquid–liquid isolation. Liquid–liquid isolation techniques such as high-speed counter-current chromatography (HSCCC) and centrifugal partition chromatography (CPC) rely on the partition of a sample between two immiscible solvents. Without a solid support matrix, the irreversible adsorption of samples can be eliminated in the stationary phase of conventional column chromatography.37 As methods of high efficiency and high recovery that are easy to scale-up, HSCCC and CPC have been used in preparative separation of ginseng saponins.38–43 Before separation with HSCCC, ginseng samples were extracted by organic solvents, and the saponin fraction was then enriched using column chromatography such as macroporous resin chromatography,39 reversed-phase C18 column,41 or medium-pressure liquid chromatography.42 A practical and effective strategy for a step-by-step selection of HSCCC conditions includes the selection of two-phase solvent systems; determination of partition coefficient (K) of analytes; preparation of the two-phase solvent system and sample solution; selection of elution mode, flow-rate and rotation speed; and on-line monitoring of the eluate.44 The selection of a suitable solvent system is the most important step in HSCCC and may be estimated as 90% of the work.37Table 2 summarizes recent applications of HSCCC to isolation of saponins from ginseng products. Ginsenosides Rb1, Rg1, Re, Rf, Rd, Rg3, Rg5, Rk1, F4, Ro and notoginsenoside R1 have been isolated by HSCCC.
Table 2 Recent applications of HSCCC to saponin isolation from ginseng products
Material Solvent systema (volume ratio) Detectionb Obtained compound Isolation efficiency c Ref.
a Abbreviations: Hex: n-hexane; BuOH: butanol; CH2Cl2: dichloromethane; MeOH: methanol; NH4OAc: ammonium acetate; iPrOH: isopropanol; CHCl3: chloroform; EtOAc: ethyl acetate. b Abbreviations: TLC: thin-layer chromatography; ELSD: evaporative light scattering detection; UV: ultraviolet. c Abbreviations: RP: reversed-phase; MPLC: medium-pressure liquid chromatography.
P. notoginseng, root Hex–n-BuOH–H2O (3[thin space (1/6-em)]:[thin space (1/6-em)]4[thin space (1/6-em)]:[thin space (1/6-em)]7) TLC Ginsenosides Rb1, Re, Rg1 and notoginsenoside R1 157, 13, 56, and 17 mg of Rb1, Re, Rg1 and R1 from 283 mg MeOH extract of five tablets 38
P. ginseng, root CH2Cl2MeOH–NH4OAc–iPrOH (6[thin space (1/6-em)]:[thin space (1/6-em)]2[thin space (1/6-em)]:[thin space (1/6-em)]4[thin space (1/6-em)]:[thin space (1/6-em)]3) ELSD Ginsenosides Rf, Rd, Re, and Rb1 10.7, 11.0, 13.4 and 13.9 mg of Rf, Rd, Re and Rb1 from 480 mg enriched fraction by macroporous resin 39
P. notoginseng, root CHCl3MeOH–2-BuOH–H2O (5[thin space (1/6-em)]:[thin space (1/6-em)]6[thin space (1/6-em)]:[thin space (1/6-em)]1[thin space (1/6-em)]:[thin space (1/6-em)]4), EtOAcnBuOH–H2O (1[thin space (1/6-em)]:[thin space (1/6-em)]1[thin space (1/6-em)]:[thin space (1/6-em)]2) ELSD Ginsenosides Rg1, Rd, Re, Rb1 and notoginsenoside R1 Not provided 40
Red P. ginseng, steamed root CH2Cl2MeOHH2OiPrOH (6[thin space (1/6-em)]:[thin space (1/6-em)]6[thin space (1/6-em)]:[thin space (1/6-em)]4[thin space (1/6-em)]:[thin space (1/6-em)]1) ELSD Ginsenosides Rg3, Rk1, Rg5 and F4 32.2, 26.6, 28.6 and 8.1 mg of Rg3, Rk1, Rg5 and F4 from 350 mg enriched fraction by RP-C18 column 41
P. ginseng, root EtOAciPrOH–0.1% formic acid in H2O (3[thin space (1/6-em)]:[thin space (1/6-em)]1[thin space (1/6-em)]:[thin space (1/6-em)]5) UV Ginsenoside Ro 61 mg Ro from 100 mg enriched sample by normal-phase MPLC 42


CPC is also a liquid–liquid partition chromatography without sorbent working in a constant-gravity field. Ginsenosides Rc, Rb1, and Re were isolated from P. quinquefolius by CPC using a two-phase solvent system of ethyl acetaten-butanolwater (1[thin space (1/6-em)]:[thin space (1/6-em)]1[thin space (1/6-em)]:[thin space (1/6-em)]2).43 Both CPC and HSCCC require the technician to be familiar with the experimental conditions and the practical separation procedure, which are unique.44

2.2.3 Recently reported ginseng saponins. Most ginseng saponins belong to a family of steroids with a four trans-ring rigid steroid skeleton. They are also referred to as ginsenosides, triterpenoid saponins or dammarane derivatives. More than 200 saponins have been isolated from ginseng plants.11 In addition to studies that focus on ginseng root, there have been studies of ginseng leaves and stems,5,45,46 flower buds,30,31,47 fruits,48,49 berries,50 and seeds.51 Because steaming or heating changes the saponin profile of ginseng products,3,52 some researchers have isolated ginseng saponins from processed roots,33,53 leaves,54 flower-buds34 and berries.55

Ginseng saponins are divided into several groups. Two major groups are the protopanaxadiol (PPD)-type saponins with sugar moieties attached to the C-3 and/or C-20, and the protopanaxatriol (PPT) group with sugar moieties at C-6 and/or at C-20. Other groups include the ocotillol-type with a five-membered epoxy ring at C-20, the oleanane-type with a nonsteroidal structure, and the dammarane type with a modified C-20 side chain.9,56 As techniques are developed for chemical purification and structural identification, novel ginseng saponins continue to be discovered. Fig. 2–4 and Table 3 show the structures of 123 new dammarane-type saponins isolated from various parts of Panax plants in the period from January 2000 to September 2010. They are placed in the order of the structure type. Compounds 4 and 91 have different structures, but both have been named ginsenoside Rh5 (Table 3). Apart from ginseng saponins, other novel compounds also have been characterized including polyacetylenes,91–95 polysaccharides,96 phenolic components,97,98 amino acids99 and alkaloids.100


Chemical structures of recently isolated dammarane-type saponins from ginseng, compounds 1–71.
Fig. 2 Chemical structures of recently isolated dammarane-type saponins from ginseng, compounds 1–71.

Chemical structures of recently isolated dammarane-type saponins from ginseng, compounds 72–95.
Fig. 3 Chemical structures of recently isolated dammarane-type saponins from ginseng, compounds 72–95.

Chemical structures of recently isolated dammarane-type saponins from ginseng, compounds 96–123.
Fig. 4 Chemical structures of recently isolated dammarane-type saponins from ginseng, compounds 96–123.
Table 3 Isolated ginseng saponins reported from 2000–2010
No. Name Formula Plant Material Ref.
1 Floralginsenoside M C53H90O22 Flower buds of P. ginseng 57
2 Floralginsenoside N C53H90O22 Flower buds of P. ginseng 57
3 Floralquinquenoside E C53H90O22 Flower buds of P. quinquefolius 35
4 Ginsenoside Rh5 C37H64O9 Roots and rhizomes of P. vietnamensis 58
5 Notoginsenoside FP1 C47H80O18 Fruit pedicels of P. notoginseng 59
6 Notoginsenoside M C48H82O19 Roots of P. notoginseng 60
7 Notoginsenoside N C48H82O19 Roots of P. notoginseng 60
8 Notoginsenoside Rw1 C46H78O17 Rhizomes of P. notoginseng 61
9 Notoginsenoside T3 C38H66O9 Acid hydrolysate roots of P. notoginseng 62
10 Notoginsenoside U C42H72O14 Roots of P. notoginseng 63
11 Quinquenoside L17 C47H80O18 Leaves and stems of P. quinquefolius 5
12 Yesanchinoside D C44H74O15 Underground part of P. japonicus 64
13 Yesanchinoside E C54H92O23 Underground part of P. japonicus 64
14 Yesanchinoside F C56H94O24 Underground part of P. japonicus 64
15 20(S)-acetylated Rg2 C44H74O14 Roots of P. quinquefolius 65
16 20(R)-acetylated Rg2 C44H74O14 Roots of P. quinquefolius 65
17 Malonylginsenoside Ra3 C62H102O30 Fresh roots of P. ginseng 66
18 Malonylnotoginsenoside R4 C62H102O30 Roots of P. ginseng 67
19 Notoginsenoside FP2 C58H98O26 Fruit pedicels of P. notoginseng 59
20 Notoginsenoside FT1 C47H80O17 Acid hydrolysate roots of P. notoginseng 68
21 Notoginsenoside L C53H90O22 Roots of P. notoginseng 60
22 Notoginsenoside O C52H88O21 Flower buds of P. notoginseng 69
23 Notoginsenoside P C52H88O21 Flower buds of P. notoginseng 69
24 Notoginsenoside Q C63H106O30 Flower buds of P. notoginseng 69
25 Notoginsenoside S C63H106O30 Flower buds of P. notoginseng 69
26 Notoginsenoside T C64H108O31 Flower buds of P. notoginseng 69
27 Quinquenoside L10 C47H80O17 Leaves and stems of P. quinquefolius 46
28 Quinquenoside L14 C47H80O17 Leaves and stems of P. quinquefolius 46
29 Yesanchinoside J C61H102O28 Underground part of P. japonicus 70
30 Floralginsenoside A C42H72O16 Flower buds of P. ginseng 71
31 Floralginsenoside C C41H70O15 Flower buds of P. ginseng 71
32 Floralginsenoside H C50H84O21 Flower buds of P. ginseng 47
33 Floralginsenoside J C48H82O20 Flower buds of P. ginseng 47
34 Floralginsenoside Ka C36H62O11 Flower buds of P. ginseng 31
35 Floralginsenoside Tc C53H90O24 Flower buds of P. ginseng 30
36 Floralquinquenoside B C42H72O15 Flower buds of P. quinquefolius 35
37 Floralquinquenoside D C42H72O15 Flower buds of P. quinquefolius 35
38 Floranotoginsenoside B C53H90O24 Flowers of P. notoginseng 72
39 Floranotoginsenoside C C53H90O24 Flowers of P. notoginseng 72
40 Ginsenoside I C48H82O20 Flower buds of P. ginseng 73
41 Ginsenoside II C48H82O20 Flower buds of P. ginseng 73
42 Ginsenoside SL1 C36H62O11 Steamed leaves of P. ginseng 74
43 Floralginsenoside B C42H72O16 Flower buds of P. ginseng 71
44 Floralginsenoside D C41H70O15 Flower buds of P. ginseng 71
45 Floralginsenoside E C42H72O15 Flower buds of P. ginseng 71
46 Floralginsenoside F C42H72O15 Flower buds of P. ginseng 71
47 Floralginsenoside G C50H84O21 Flower buds of P. ginseng 47
48 Floralginsenoside I C48H82O20 Flower buds of P. ginseng 47
49 Floralginsenoside K C48H82O21 Flower buds of P. ginseng 47
50 Floralginsenoside O C53H90O24 Flower buds of P. ginseng 57
51 Floralginsenoside P C53H90O23 Flower buds of P. ginseng 57
52 Floralquinquenoside A C36H62O11 Flower buds of P. quinquefolius 35
53 Floralquinquenoside C C42H72O15 Flower buds of P. quinquefolius 35
54 Ginsenoside Rh6 C36H62O11 Leaves of P. ginseng 32
55 Floralginsenoside La C48H82O19 Flower buds of P. ginseng 47
56 Floralginsenoside Lb C48H82O19 Flower buds of P. ginseng 47
57 Floranotoginsenoside D C53H90O23 Flowers of P. notoginseng 72
58 Ginsenoside Rg7 C36H60O9 Leaves of P. ginseng 32
59 Notopanaxoside A C36H62O10 Roots of P. notoginseng 75
60 Notoginsenoside FT3 C47H80O18 Acid hydrolysate roots of P. notoginseng 68
61 Floranotoginsenoside A C53H90O23 Flowers of P. notoginseng 72
62 Ginsenoside ST2 C36H62O10 Steamed leaves of P. ginseng 54
63 Notoginsenoside Rw2 C41H70O14 Rhizomes of P. notoginseng 61
64 Notoginsenoside ST5 C47H80O18 Steamed roots of P. notoginseng 33
65 Yesanchinoside H C53H90O23 Underground part of P. japonicus 70
66 Ginsenoside Ki C36H62O10 Leaves of P. ginseng 76
67 Ginsenoside Km C36H62O10 Leaves of P. ginseng 76
68 Quinquenoside L2 C48H82O19 Leaves and stems of P. quinquefolius 77
69 Dammar-25(26)-ene-3,6,12,20,22,24-hexanol C30H52O6 Leaves of P. ginseng 78
70 Floralginsenoside Kb C45H76O19 Flower buds of P. ginseng 31
71 Floralginsenoside Kc C45H76O20 Flower buds of P. ginseng 31
72 Floralginsenoside Ta C36H60O10 Flower buds of P. ginseng 30
73 Vina-ginsenoside R25 C42H70O15 Roots and rhizomes of P. vietnamensis 58
74 Floralginsenoside Tb C35H62O11 Flower buds of P. ginseng 30
75 Quinquenoside L9 C42H74O15 Leaves and stems of P. quinquefolius 79
76 Quinquenoside L16 C54H94O25 Leaves and stems of P. quinquefolius 46
77 25-OCH3-PPD C31H56O4 Leaves of P. notoginseng 80
78 25-OH-PPD C30H54O4 Fruits of P. ginseng 48
79 25-OH-PPT C30H54O5 Fruits of P. ginseng 48
80 Notoginsenoside FT2 C47H82O18 Acid hydrolysate roots of P. notoginseng 68
81 Notoginsenoside T4 C36H62O11 Acid hydrolysate roots of P. notoginseng 62
82 Quinquenoside L1 C48H80O18 Leaves and stems of P. quinquefolius 77
83 Quinquefoloside La C54H92O23 Leaves of P. quinquefolius 45
84 Quinquefoloside Lc C54H92O23 Leaves of P. quinquefolius 81
85 Dammar-(E)-20(22)-ene-3,12,25-triol C30H52O3 Acid hydrolysate roots of P. ginseng 82
86 Notoginsenoside ST1 C36H62O10 Steamed roots of P. notoginseng 33
87 Ginsenoside Rg6 C42H70O12 Stem-leaves of P. ginseng 83
88 Ginsenoside Rs4 C42H70O12 Steamed roots of P. notoginseng 84
89 Ginsenoside Rs6 C42H70O12 Steamed roots of P. notoginseng 84
90 Isoginsenoside Rh3 C36H60O7 Fruits of P. ginseng 49
91 Ginsenoside Rh5 C36H60O9 Leaves of P. ginseng 32
92 Ginsenoside SL2 C42H70O14 Steamed leaves of P. ginseng 74
93 Ginsenoside ST1 C36H60O10 Steamed leaves of P. ginseng 54
94 Notoginsenoside ST2 C43H74O15 Steamed roots of P. notoginseng 33
95 Notoginsenoside ST3 C43H74O15 Steamed roots of P. notoginseng 33
96 Ginsenoside Rg8 C42H70O12 Roots of P. quinquefolius 85
97 Notoginsenoside T1 C36H60O10 Acid hydrolysate roots of P. notoginseng 62
98 Notoginsenoside T2 C36H62O10 Acid hydrolysate roots of P. notoginseng 62
99 12,23-Epoxyginsenoside Rg1 C42H70O14 Leaves of P. ginseng 86
100 Ginsenoside Rh9 C36H60O9 Leaves of P. ginseng 32
101 Quinquefoloside-Lb C53H88O22 Leaves of P. quinquefolius 45
102 Ginsenoside Rk1 C42H70O12 Processed roots of P. ginseng 36
103 Ginsenoside Rk2 C36H60O7 Processed roots of P. ginseng 36
104 Ginsenoside Rk3 C36H60O8 Processed roots of P. ginseng 36
105 Ginsenoside Rs5 C38H62O9 Steamed roots of P. notoginseng 84
106 Ginsenoside Rs7 C38H62O9 Steamed roots of P. notoginseng 84
107 Notoginsenoside T5 C41H68O12 Acid hydrolysate roots of P. notoginseng 62
108 Ginsenoside Rz1 C42H70O12 Steamed roots of P. notoginseng 53
109 Ginsenoside SL3 C42H70O14 Steamed leaves of P. ginseng 74
110 Ginsenoside Rh8 C36H60O9 Leaves of P. ginseng 32
111 Ginsenoside Rh7 C36H60O9 Leaves of P. ginseng 32
112 Yesanchinoside G C53H88O23 Underground part of P. japonicus 70
113 Yesanchinoside I C59H100O26 Underground part of P. japonicus 70
114 Hexanordammaran C24H40O4 Leaves of P. ginseng 87
115 Notoginsenoside R10 C30H50O9 Steamed leaves of P. ginseng 88
116 Yesanchinoside A C44H74O16 Underground part of P. japonicus 64
117 Yesanchinoside B C48H82O20 Underground part of P. japonicus 64
118 Yesanchinoside C C47H80O19 Underground part of P. japonicus 64
119 Panaxadione C30H48O5 Seeds of P. ginseng 51
120 Polyacetyleneginsenoside Ro C65H100O21 Roots of P. ginseng 89
121 Isodehydroprotopanaxatriol C30H50O3 Acid hydrolysate roots of P. ginseng 90
122 20,25-Epoxydammaran-2-en-6,12-diol C30H50O3 Acid hydrolysate roots of P. ginseng 90
123 3-Methyl-28-nordammaran-2-en-6,12-diol C30H50O3 Acid hydrolysate roots of P. ginseng 90


2.3 Knockout technologies

The constituents and functions of ginseng are studied by isolation-to-bioassay or bioassay-guided isolation.1 To prove whether the obtained constituents are active compounds of the extract, it is necessary to prepare an extract in which the assumed compound(s) is removed,101 called a “knockout” extract. The “knockout” concept has been widespread in genetic engineering for pharmacologic studies since 1989.102 In bioactivity comparisons, if a knockout extract has low biological activity compared with the activity of the original extract, the eliminated constituents can be considered bioactive compounds.101 Therefore, one goal of studies is to validate methods for preparing knockout extracts. Chemical chromatography and immunoaffinity chromatography have been introduced for this purpose.
2.3.1 Chemical chromatography. Some knockout extracts are prepared by column chromatography. For example, ginsenoside Re was believed to be the antihyperglycemic active compound in P. quinquefolius berry extract.103 To prepare the Rb1 knockout extract, ginseng berry extract was loaded onto an HP-20 chromatographic column. The column was eluted with water and then aqueous ethanol. The aqueous ethanol eluate portion was separated by reversed-phase preparative high-performance liquid chromatography (HPLC) to give three fractions: a water fraction, an Rb1 fraction, and a fraction containing other saponins. The Rb1 fraction was removed, and the water fraction and saponin portion combined to prepare the Rb1 knockout extract.

To improve efficiency, Liu et al.101 developed an on-line chromatography technology to prepare the knockout extract. As shown in Fig. 5, an HPLC technique was used to separate constituents in a herb extract, ensuring target compounds achieved baseline separation from adjacent peaks. Then a six-port valve-switching unit ‘cut and captured’ target compounds according to the chromatographic retention times. Ingredients were thus divided into two parts: the target compounds and others without the target. Based on valve-switching, the main bioactive and characteristic components (or a combination of them) can be obtained. Fig. 5 also shows an application example with ginseng. With HPLC, ginseng extract underwent baseline separation of its major components.104 Through the valve switching technique, the original extract, the Re and Rb1 knockout extract, and knockout compounds (Re + Rb1) can be obtained. A comparison of the bioactivities of the samples shows the overall efficacy of target peaks, the bioactive contribution of each component, and the interaction of multiple components.


Online chromatographic knockout technique, modified from ref. 101. An example is shown for P. quinquefolius, with ginsenosides Re and Rb1 being knocked out simultaneously.
Fig. 5 Online chromatographic knockout technique, modified from ref. 101. An example is shown for P. quinquefolius, with ginsenosides Re and Rb1 being knocked out simultaneously.
2.3.2 Immunoaffinity chromatography. Immunoaffinity chromatography is a powerful technique to isolate and concentrate minor components of interest from complex mixtures.105,106 Its selectivity is derived from the use of an immobilized binding protein on a suitable solid-phase support. In a continuous effort to produce monoclonal antibodies (MAbs) for naturally occurring bioactive compounds, Shoyama et al. prepared anti-ginsenoside Rb1,107 Rg1,108 Rd,109 and Re MAbs.110 The antibody specificity was tested by determining the cross-reactivities of the MAbs with structurally similar compounds by competitive enzyme-linked immunosorbent assay (ELISA).111–113

Fig. 6 shows a protocol for preparing ginsenoside Rb1 knockout ginseng extract.113–115 The first step is to obtain anti-ginsenoside Rb1 MAbs from mice through synthesis of antigen conjugates, immunization, hybridization and purification. Since anti-ginsenoside Rb1 MAbs have a sugar chain in the molecule, the second step is to produce aldehyde groups through oxidative cleavage by sodium periodide (NaIO4).115,116 The oxidized product is treated with agarose gels conjugated with a hydrazine group to give hydrazone. The immunoaffinity column is produced with an affinity gel combined with MAbs.110,114 After method validation, ginseng extract is charged to the column, washed with phosphate buffer and then eluted with acetate buffer and 20% methanol to give pure ginsenoside Rb1. All washed buffer solutions are collected and deionized. The water solution is concentrated and subjected to thin-layer chromatography (TLC) or HPLC analysis. This immunoaffinity chromatographic method can establish one-step preparation of Rb1 knockout ginseng extract.


Immunoaffinity chromatographic method for preparation of Rb1 knockout extract from P. ginseng extract, modified from ref. 114. The red spots in the top right part of the graphic indicate Rb1. Lines 1, 2, and 3 in the thin-layer chromatography (TLC) plate in the bottom right part of the graphic indicate crude extract, knockout extract, and purified Rb1, respectively.
Fig. 6 Immunoaffinity chromatographic method for preparation of Rb1 knockout extract from P. ginseng extract, modified from ref. 114. The red spots in the top right part of the graphic indicate Rb1. Lines 1, 2, and 3 in the thin-layer chromatography (TLC) plate in the bottom right part of the graphic indicate crude extract, knockout extract, and purified Rb1, respectively.

Compared with chemical chromatography for knockout, immunoaffinity chromatography enhanced analytical selectivity, decreased the number of tedious steps in sample preparation, and improved sampling loading volume. Additionally, the run-time of chromatographic separations and optimal conditions is reduced. However, immunoaffinity chromatography has some limitations, namely complicated MAbs and column preparation, non-commercial availability of MAbs, and instability of the immunoaffinity columns.

3 Analytical advances

In this section, we emphasize emerging techniques, general trends, and recent advances for the analysis of ginseng. Some methods highlighted are (for separation): high-performance thin-layer chromatography (HPTLC); hydrophilic interaction chromatography (HILIC); ultrahigh-pressure liquid chromatography (UPLC); multidimensional chromatography; and on-line concentration electrokinetic chromatography; and (for detection) advanced mass spectrometry (MS), i.e. quadrupole (Q)-MS, ion trap (IT)-MS, time-of-flight (TOF)-MS instruments, and more recently, Q-IT, IT-TOF and Q-TOF.

3.1 Direct profiling technologies

With the development of chemometrics and informatics in multivariate statistical analysis, spectrometric and spectroscopic methods are frequently applied to the profiling of crude ginseng extracts without prior separation. Profiling without hyphenation technologies is useful for rapid evaluation of the composition, authentication of the ginseng plant species, quality control, and metabolomics.117 The major techniques are nuclear magnetic resonance (NMR), infrared (IR) and Raman spectroscopy.
3.1.1 Nuclear magnetic resonance (NMR) spectroscopy. The profiling of crude ginseng extracts can be performed by direct NMR analysis. This method, which requires limited sample preparation, is simple, comprehensive and reproducible. Direct NMR profiling is applied in metabolomic fingerprinting for quality control and authentication of different ginseng species, ages, and origins. With the aid of chemical shifts and coupling constants in the 1H NMR spectrum, constituents in ginseng can be identified. The concentration of each compound in the sample can be calculated from integration of the signals in the spectra without calibration curves.118 Multivariate data analysis sorts the discriminating signals from the complex spectra.119

There are several reports on the discrimination of ginsengs by NMR profiling. Sugar and methyl signals in ginseng saponins obtained from 1H-NMR with principal component analysis (PCA) and cluster analysis can differentiate geographical origins and ages of P. ginseng roots.120,121 Ginseng saponins, saccharides, amino acids, fumaric acid and inositol were screened as important markers by two-dimensional (2-D) NMR with PCA to discriminate between P. ginseng and P. quinquefolius preparations and white and red ginseng root.122

Direct NMR profiling is powerful for recording changes in composition within a ginseng mixture. One of the main drawbacks of this method is its low sensitivity – it is not well suited for detecting minor changes in composition.

3.1.2 Infrared (IR) and Raman spectroscopy. IR is an important tool in organic chemistry to assess a compound's purity and its structure. Pattern recognition with IR has been applied to analyze ginseng composition and for species authentication and quality control. The main advantages of IR over the traditional chemical and chromatographic methods are its simplicity in routine operations. It is non-destructive and requires less sample preparation. One drawback of IR is the calibration step for quantitative analysis, which requires analysis of a set of samples covering the spectral variability in the sample.

Based on a unique IR spectral fingerprint in the 2000–600 cm−1 region, Yap et al. described a “2-6PC rule” for categorizing ginseng products to authenticate them from morphological adulterations123 and to distinguish among P. ginseng, P. quinquefolius and P. notoginseng.124 Near-infrared spectroscopy was used to determine the total sugar content in ginsengs.125 Fourier transform infrared (FTIR) spectroscopy with support vector machine and wavelet transform technology identified the cultivation area of ginsengs126 and differentiate P. quinquefolius from P. ginseng.127 In a recent study, 2-D FTIR and 2-D IR correlation spectroscopy provided colorful maps and dynamic structural information about chemical components to authenticate P. ginseng, P. quinquefolius and P. notoginseng,128 to differentiate cultivated from wild ginseng,129 to discriminate between the different geographical sources of P. quinquefolius,130 and to evaluate the different ages of ginseng samples.131

Raman spectroscopy with pattern recognition has been used to classify ginsengs according to species and processing method132 and to investigate polyacetylenes in P. quinquefolius roots.133

3.2 Separation-based methods

One emerging trend has been combining techniques, which have been useful for profiling the composition of ginseng samples. Two major steps are involved – separation and detection. Separation of ginseng saponins is performed by TLC or HPTLC; HPLC, HILIC or UPLC; gas chromatography (GC); multidimensional chromatography; or electrophoresis (CE). Among these techniques, liquid chromatography is used most frequently. A comparison of different separation modes for ginseng analysis is given in Table 4.
Table 4 Separation modes employed in ginseng analysisa
Method Advantages Limitations Ref.
a Abbreviations: HPTLC: high-performance thin layer chromatography; GC: gas chromatography; HPLC: high-performance liquid chromatography; HILIC: hydrophilic interaction liquid chromatography; UPLC: ultra-performance liquid chromatography; CE: electrophoresis; 2-D: two-dimensional; ESI: electrospray ionization; MS: mass spectrometry.
TLC/HPTLC Easy to use Low efficiency in separation 134,135
Versatility Low reproducibility
Low cost Low accuracy in quantification
 
GC High sensitivity Limited to volatile compounds 91,136–138
High resolution Time consumption in derivation
High speed Thermal instability
 
HPLC Widely available Extended analytical time 28,74,104,121,139–141
Relatively easy to use Relatively high cost
High accuracy and precision
 
HILIC Suitable for hydrophilic molecules Peak broadening 142–145
Beneficial to ESI process Short column lifetime
Weak interaction with stationary phase
 
UPLC Highly efficient separation Back-pressure increase 146–148
Reduced analytical time Dedicated instrumentation
Reduced solvent consumption
 
CE Short analytical time Low reproducibility 149–152
Easy to automate Insufficient detection sensitivity
Environmentally friendly Complicated interfacing to MS
 
2-D chromatography Powerful separation ability Possibility of artifact formation 136,153
High peak capacity Extended analysis time
Excellent detection sensitivity Limitations in mobile phase


3.2.1 Thin-layer chromatography (TLC) and high-performance TLC (HPTLC). For a considerable time, TLC or planar chromatography (PC) was called the “Cinderella” or the “princess” of chromatographic techniques. The main advantages of TLC are its versatility, speed, flexibility and low cost. It is used for tests of purity, quick qualification or species identification in pharmacopoeias and the pharmaceutical industry. To avoid the harmful reagents noted in the 14th edition of the Japanese Pharmacopoeia, Ohno et al.135 developed a reversed-phase TLC using a sodium sulfateacetonitrilemethanol solution instead of a chloroformmethanolwater solution as the developing solvent with Rg1 as the marker. Detection of ginseng saponins by TLC is usually achieved by sulfuric acid alone or its mixture with aromatic aldehydes like vanillin or anisaldehyde.

Many TLC procedures have been carried out under high-performance conditions (HPTLC). In HPTLC, the plates are precoated for the stationary phase with a mean particle size of 5 μm for better separation and reproducibility. HPTLC was developed for differentiation of the ginseng species. A fingerprint pattern with HPTLC discriminates white and red P. ginseng, P. quinquefolius and P. notoginseng.154,155 With the introduction of densitometry, TLC also has become a useful tool for the quantitative analysis of ginseng saponins. A typical application was performed by Vanhaelen-Fastré et al.,156 who used HPTLC for simultaneous quantification of six major ginseng saponins in P. ginseng. Kevers et al.157 developed an automatic HPTLC sampler and scanner that determines ginseng saponins and identifies the different species and sources. The major disadvantages of TLC and HPTLC are low accuracy and reproducibility.

3.2.2 Gas chromatography (GC). GC is sensitive for detecting volatile chemical compounds or non-volatile compounds readily derivatized.1 Most GC uses capillary columns (commonly fused-silica, 30 m × 0.25 mm × 0.25 μm). For detection, the flame ionization detector has remained popular. Structural information and selectivity available from mass spectrometry (MS) has made the combination of GC and MS more effective. The strategy for GC analysis of ginseng saponins is to hydrolyze the saponins before trimethylsilyl-derivatization of their genuine aglycones to simplify and improve sensitivity.11

GC has been used for analysis of volatile organic compounds (e.g., oils).136,158 Sample preparation is the crucial first step in GC analysis of volatile compounds in ginseng. Novel sample preparation techniques have been established to extract volatile compounds from the matrix, including headspace solid-phase microextraction137 and solvent-free solid injection based on direct vaporization.158 Another important use of GC has been to analyze multiresidue pesticides and their metabolites in ginseng samples. There are reports of methods that describe the screening of pesticides in dried ginseng powders, extracts and preparations.151,159,160 To make methods faster, automated, and cost-efficient, GC has been combined with high resolution time-of-flight (TOF) MS.160

GC can also determine other active ingredients. There are methods for measuring semi-volatile components such as sesquiterpenes and polyacetylenes91,153 and for quantifying phenolic constituents after derivatization to their trimethylsilyl derivatives.161 GC is also a powerful tool to test endogenous metabonomics in biological samples for ginseng's therapeutic effects and toxicity.138

As has been the general trend for a number of years, GC is decreasing in popularity, as reflected by the number of new procedures being reported. Since the complex patterns of the different types of ginseng saponins cannot be evaluated with GC, it is rarely used in ginseng studies.

3.2.3 High-performance liquid chromatography (HPLC), hydrophilic interaction LC (HILIC) and ultra-performance LC (UPLC). HPLC continues to be the most used technique in ginseng research because it is low-cost, readily available, and easy to use. Typically, HPLC is performed under reversed-phase conditions with standard 150 or 250 mm × 4.6 mm columns packed with 5-μm porous silica-based C18 sorbents. In the mobile phase, acetonitrile and water with buffer are used, and a gradient elution program is optimized. Although HPLC may be considered a mature technology, advances continue to improve the separation capability and productivity of the method and sample analysis.

HILIC has been gaining interest in the last few years as an alternative to HPLC.142–145 HILIC is especially useful for separation of polar and hydrophilic analytes as well as polar metabolites in biological samples. Separations are carried out in the hydrophilic stationary phase using isocratic hydro-organic eluents that contain high levels of the organic component. Retention times increase with the hydrophilicity of the analytes. When combined with MS, high organic content in the mobile phase benefits the electrospray ionization process. The influence of column temperature and the choice of eluent components such as buffer are key to separation efficiency and selectivity.145 In HILIC, a silica column (50 × 2.1 mm, 3 μm) was used to analyze dencichine (a neuro-excitatory non-protein amino acid) in different ginseng species with a run time of approximately 5 min. Dencichine was not retained by any of the reversed-phase packing.142 HILIC using diols,143 or polyvinyl alcohol-bonded,145,162 or polyamine-bonded144 stationary phases was developed for the separation and quantitative determination of ginseng saponins in ginseng preparations. Shorter analysis time (15–30 min compared with more than 60 min in RP-HPLC) and better resolution were achieved. The elution window for the analysis of compounds that are difficult to separate can be broadened by the combination of HILIC with conventional HPLC.163

UPLC, which uses short columns packed with particles less than 2 μm in size, has emerged as a powerful method in many laboratories. Chromatographic separation is 5–10 times faster than HPLC, without a reduction in resolution.146,164,165 Existing HPLC conditions can be directly transferred to UPLC. Combining UPLC with an MS detector fulfils key requirements in terms of rapidity, sensitivity, selectivity, and peak-assignment certainty for the analysis of analytes at low concentrations in complex matrices.166 UPLC has been popular for rapid separation of ginseng saponins in metabolomic fingerprinting,146–148 quality control,167 and biological samples.168,169 A comparison was made of the performance of UPLC on a 100 × 2.1 mm, 1.7 μm column and of HPLC on a 250 × 4.6 mm, 5.0 μm column. Twenty-five ginseng saponins were identified within a 20-min run time with UPLC, while fewer markers were recognized by HPLC within 80 min.148 At present, the major limitation of UPLC is the cost of dedicated equipment and consumables, keeping the instrument more of a research than a routine tool. To provide a more economical alternative, a simplified approach was recently introduced, in which a modified conventional HPLC system was connected directly with short columns to reduce time for analysis of complex herbal medicines.170,171 Direct connection offers opportunities for laboratories looking to improve separation without upgrading an existing HPLC system to an UPLC system.

3.2.4 Micellar electrokinetic chromatography (MEKC) and microemulsion EKC (MEEKC). Ginseng saponins do not have charges, so capillary zone electrophoresis (CZE) is not applicable. Therefore, micellar electrokinetic chromatography (MEKC) and microemulsion electrokinetic chromatography (MEEKC) are used for the separation of neutral ginseng saponins. MEKC is efficient and fast, and running buffer composition can be tuned or changed to select separation.172 Compared to MEKC, the presence of a water-immiscible oil phase in the microemulsion droplets of MEEKC gives rise to some special properties, such as enhanced capacity for solubilization and an enlarged migration window.173

In general, analysis of ginseng saponins with CE can be optimized by sampling and concentrating the ginseng samples, buffer additives, surface modifications and detections.174 The concentration sensitivity of MEKC and MEEKC is poor because the best path is short and only a small volume of sample can be injected. To improve the sensitivity of MEKC, on-line concentration techniques with stacking procedures have been developed. Wang et al.175 developed a field-enhanced sample injection with reverse migrating micelles to separate and concentrate ten ginseng saponins in P. notoginseng on-line. Ginseng saponins achieved separation within 35 min, shorter than the 65 min by HPLC. Sensitivity was one magnitude higher than with the method by Glöckl et al.,149 enabling two minor ginsenosides, Rh1 and Rg2, to be determined.

Cao et al.152 used on-line dual sweeping based on borate complexation and enhancement of the organic solvent field enhancement for the preconcentration of ginsenosides Rf, Rg1, and Re in MEKC with nonionic Brij-35 micelles. Subsequently, they established a complex formation and acetonitrile sweeping technique in non-ionic MEEKC for ginsenosides Rf, Rb2 and Re using a Brij-35 microemulsion.151 Pressure and electrokinetic injections of long sample plugs were also presented for simultaneous stacking of notoginsenoside R1, and ginsenosides Rg1, Rf, Rh1, Rd, Rg3 in MEEKC.150 Improvements in sensitivity were in the range 50- to 200-fold.

In spite of novel CE techniques, the applications of CE for analysis of ginseng are limited. One example uses a CE microchip coupled with a polymerase chain reaction (PCR) analysis of short tandem repeats for rapid authentication of ginseng species.174 Most applications are for protein analysis to decipher ginseng's biological mechanisms.

3.2.5 Multidimensional chromatography. The separation power of traditional one-dimensional techniques is sometimes inadequate for complex samples. In such a case, multidimensional chromatography is used, and most multidimensional separations are 2-D. In 2-D separation, a sample is first separated by one method, and then the separated compounds are subjected to an additional independent method. For complex samples like ginseng extracts, multidimensional chromatography offers more powerful separation ability, higher peak capacity and detection sensitivity than single-dimension chromatography. Multidimensional chromatography can be easily realized by the use of GC and TLC. The realization of multidimensionality in LC is difficult because of problems in switching the mobile phase from one system to another.176

Shellie et al.153 developed comprehensive 2-D GC (GC[thin space (1/6-em)]×[thin space (1/6-em)]GC) to identify qualitative differences between Panax species extracts. GC[thin space (1/6-em)]×[thin space (1/6-em)]GC revealed the presence of numerous common components and possible species-specific components. Qiu et al.136 described a comprehensive GC[thin space (1/6-em)]×[thin space (1/6-em)]GC technique to analyze the chemical composition of volatile oil in P. ginseng at different ages. This method for PCA classifies and differentiates ginseng samples at different ages.

With 2-D chromatography, artifacts may form, polar and nonvolatile solvents may be unavailable in the mobile phase, and analysis times are long.

3.3 Hyphenated detection-based methods

After sample separation, the next step is to detect the target compounds. Hyphenated detection-based methods include ultraviolet (UV) or diode-array detection (DAD), evaporative light scattering detection (ELSD), charged aerosol detection (CAD), MS and ELISA. Different detectors can be combined to obtain complementary information in one run, such as DAD-ELSD and DAD-MS. Table 5 compares the advantages and limitations of the various methods.
Table 5 Characterization of detection methods for ginseng analysis
Methoda Establishment Sensitivity Structure Linear range Operation Precision Accuracy Cost
a Abbreviations: UV/DAD: ultraviolet or diode-array detection; ELSD: evaporative light scattering detection; CAD: charged aerosol detection; MS: mass spectrometry; ELISA: enzyme-linked immunosorbent assay.
UV/DAD Very easy Moderate Possible Very wide Simple Fairly good Very good Very low
ELSD Easy Low No Narrow Fair Good Good Low
CAD Easy Moderate No Wide Fair Good Good Low
MS Not easy High Powerful Narrow Not simple Low Low High
ELISA Very complex Very high Possible Very narrow Simple Low Low Low


3.3.1 Ultraviolet (UV) or diode-array detection (DAD). Analysis by UV continues to be the workhorse of the pharmaceutical laboratory. It is available in laboratories, inexpensive, and easy to use. Because of weak UV absorption by ginseng saponins, detection is usually set at 198–205 nm.3,50 Weak absorption affects the sensitivity because of the interference of other compounds that absorb short wavelengths. The choice of mobile phase and modifiers is also limited. UV detection systems are combined with LC and CE for analysis of ginseng saponins. Instead of UV, DAD has become popular in many laboratories, since it monitors several wavelengths and provides “on-the-fly” spectra at once which maximizes sensitivity and assures peak purity.177 UV and DAD are used to quantify ginseng saponins in extract and biofluids because of their accuracy and precision. In a recent trend, UV/DAD has been connected on-line with MS. UV/DADMS techniques yield information for rapid identification and quantification of constituents in ginseng.
3.3.2 Evaporative light scattering detection (ELSD). The main problem encountered in performing UV analyses of ginseng is the relatively high level of baseline noise.13,55 ELSD is a universal, non-specific detection system that provides a stable baseline even with gradient elution.140 Furthermore, with volatile mobile-phase modifiers, such as HCOOH and CH3COOH, better selectivity can be obtained.18 With ELSD, non-chromophoric ginseng saponins such as 24(R)-pseudoginsenoside F11 can be analyzed.140 Parameters to be settled for ELSD are the flow rate of the nebulizer gas and the drift tube temperature.178,179 The sensitivity of ELSD was found to be 5 times lower than that obtained with UV, with a minimum detectable concentration of 100–200 ng of ginseng saponins on the column.28,140

An HPLCELSD method has been developed for simultaneous monitoring of 12 ginseng saponins in different parts of P. quinquefolius,28 14 saponins in red P. ginseng,180 and 19 saponins in black ginseng.17 Wan et al.18 described the chemical characteristics of three medicinal plants of the Panax genus by HPLCELSD. Quantitative evaluation of ginseng saponins using both UV and ELSD in P. quinquefolius showed similar results, except that UV could not be applied to detect pseudoginsenoside F11.140,181 However, in light of its better sensitivity, easier handling and greater availability, UV is still recommended for the routine analysis of ginseng samples.

3.3.3 Charged aerosol detection (CAD). CAD has been introduced as an alternative to ELSD for detection of non-UV and weakly UV-active compounds and for UV-absorbing compounds in the absence of standards. Both CAD and ELSD depend on mass, and the response generated does not depend on the spectral or physicochemical properties of the analyte. With a detection technique that generates universal response factors, there is potential for a single, universal standard for calibration against which all other compounds or impurities can be qualified.182 The main advantages of CAD are better sensitivity than an ELSD system, wider dynamic range, ease of use, and constancy of response factors.

Bai et al.183 established an HPLCCAD method for quantitative analysis of notoginsenoside R1, ginsenosides Rg1, Re, Rb1, Rg2, Rh1, and Rd in 30 batches of P. notoginseng samples. The sensitivity of CAD was higher than that of ELSD or UV. Wang et al.184 compared LCCAD, LCELSD and LC–UV methods to determine ginsenosides Rg1, Re, Rb1, Rc, Rb2, Rb3 and Rd in P. ginseng. CAD had higher sensitivity than UV and ELSD, and better linearity and reproducibility than ELSD.

The limitations of CAD are the need for volatile buffers, non-volatility of analytes, and dependence on the changes in the eluent composition for a response.

3.3.4 Mass spectrometry (MS) and multi-stage MS (MS/MS). An emerging instrumental trend has been the application of MS and its hyphenation with chromatographic separation techniques. In ginseng and ginseng-treated samples, the most used of hyphenated techniques is HPLCMS, and to a lesser extent, GCMS. In a typical MS procedure, two key modules are the ion source, which can convert sample molecules in the gas phase into ions or move ions from the solution into the gas phase; and the mass analyzer, which sorts the ions by their masses. Several different ion sources are used depending on the information needed. Atmospheric pressure chemical ionization (APCI)185–187 and electrospray ionization (ESI),146,188–192 also known as “soft ionization,” are the most frequently used with LC in ginseng analysis. ESI is used more than APCI. Electron ionization or “hard ionization,” is the most commonly used GCMS technique.193

Four types of mass analyzer are used for analysis of ginseng and ginseng-related samples, i.e., quadrupole (Q),160 ion trap (IT),194 time-of-flight (TOF),147,148,164,195–197 and Fourier transform ion cyclotron resonance (FT-ICR).198,199 Quadrupoles tend to be the simplest and least expensive mass analyzers. Quadrupole mass analyzers can operate in scanning mode or selected ion monitoring (SIM) mode. Because of the scan mode's low sensitivity, the SIM mode is used for quantifying and monitoring target compounds. The SIM mode, which provides information on a few ions, is not suited for screening unknown compounds. Ion traps are also relatively inexpensive. Compared with losses with quadrupole, sensitivity losses during the full-scan mode can be avoided. An advantage of ion traps is that multiple stages of mass spectrometry can be performed without additional mass analyzers. Time-of-flight mass analyzers measure mass accurately and have high resolution and full-scan mass range. Accurate mass measurement, which gives the elemental composition of obtained ions, facilitates non-target component identification. Compared with quadrupole and ion trap, TOF is more expensive and has a narrower linear range for quantification. Like ion traps, FT-ICR mass analyzers can process mass reaction in multiple stages without additional analyzers. Like TOF, FT-ICR analyzers also have a wide mass range and a high mass resolution. They are, however, the most expensive of the mass analyzers.

Multi-stage MS (also called tandem MS or MS/MS or MSn) combines the different designs of mass analyzers. MS/MS gives advanced structural information and has the sensitivity, specificity, and versatility. Triple quadrupole (QqQ),200 quadrupole ion trap (Q-IT),194,201 quadrupole time-of-fight (Q-TOF),164,196 or ion trap time-of-flight (IT-TOF)202,203 instruments are used for ginseng analysis. Hybrid systems such as Q-TOF-MS and IT-TOF-MS, which provide fragmentation information together with accurate mass measurements of product ions, are powerful tools for structural analysis. In MS/MS, collision-induced dissociation (CID) between analyte ions and neutral molecules forms fragment ions. Voltages are adjusted to increase collision energy to produce abundant fragmentation for predicting the structure of compounds. Another advantage of multi-stage MS is that nonanalyte ions are discarded in the first stage and thus sample cleanup is less. Multi-stage MS, however, is extremely expensive, which makes it impractical for most laboratories.

CID can also be achieved in single-stage mass spectrometers (commonly called in-source CID). The advantage of performing CID in single-stage instruments is simplicity and relatively low cost.204 One application is in-source CID in single-stage TOF-MS.205 The fragmentor in TOF-MS is crucial for efficient transmission of the ions to obtain the best balance between sensitivity and fragmentation.206,207 With dynamic adjustment of the fragmentor from gentle ionization to intense fragmentation, molecular ions and abundant fragment ions are readily observed for unambiguous identification with similar or better performance than tandem MS.170,171 The disadvantage of CID in single-stage MS is that a specific precursor ion cannot be selected, so the source of product ions cannot be determined.

One limitation of MS-based methods is the inherent lack of ionization of certain components. Another is that LCMS ionization is instrument-dependent, making it difficult to construct standard mass spectrum libraries for ginseng saponins. Equipment, especially for TOF-MS, must be calibrated accurately and regularly. As with most advanced technologies, generating useful and reliable data depend on the experience and the skill of the operators.

3.3.5 Enzyme-linked immunosorbent assay (ELISA). ELISA is a biochemical test that measures the concentration of a compound using the reaction of an antibody or antibodies to its antigen. MAbs are often used. ELISA combines the advantages of the high specificity of antibodies and the great sensitivity of simple enzyme assays. Tanaka et al.114,208 developed a new immuno-staining ELISA method called Eastern blotting. In their studies, ginseng extract was first subject to a silica gel TLC plate using developing solvents. After separation, the TLC plate was transferred to a polyvinylidene fluoride membrane or a polyethersulfone membrane. Then the membrane was treated by NaIO4 and a buffer containing a carrier protein such as bovine serum albumin (BSA). The sugar moieties in ginseng saponins are oxidized by NaIO4 to produce dialdehydes. These dialdehydes react with the amino groups of lysine and/or arginine of the BSA that binds strongly to the adsorbent membrane. The membrane was further treated by a MAb and then a secondary antibody modified by enzyme. The aglycon part of the ginsenoside molecule bound to the corresponding MAb, enabling visualization by the enzyme-labeled specific antibody. Finally, a substrate was added for staining, and NIH imaging software was used for quantification. The double staining of Eastern blotting for ginseng saponins can indicate possible structures for unknown saponins, depending on the staining color related to the aglycone and the Rf value.

ELISA has been applied for single or simultaneous quantification of ginsenosides Re, Rb1, Rg1, Rd and Rc in various ginseng extracts.109,110,116,208,209 Eastern blotting has been used to identify chikusetsusaponins III and IV in P. japonicus.114 ELISA suffers from the complex preparation of MAbs, strong cross-reactivity caused by some structurally similar constituents, and narrow dynamic linear range for quantification.

4 Applications and challenges

As one of the most widely studied medicinal plants and herbs during the last decade, a great number of scientists have contributed to developing methods for analyzing ginseng. Advances in isolation and analysis technology have improved analysis times, efficiency, and method development. Application of these methods has been made to various aspects of ginseng and ginseng-related samples. In this section, we review species differentiation, quality assessment, and interaction with biological systems. Challenges in studying ginseng are also included.

4.1 Species authentication

Ginseng grows or is cultivated in different geographical regions. Although ginsengs are closely related taxonomically, and contain similar chemical constituents, they differ in their effects on symptoms in people. In the herbal market, adulterants are sometimes part of samples.128,210 Because the roots of ginsengs are similar in appearance and many commercial ginseng products are sold as powder or shredded slices, identification of the origins of a product is not easy. Some new methods have been developed to differentiate ginseng species, mainly through chemical profiling or molecular markers.
4.1.1 Chemical profiling. Different ginseng species, ages, and geographical sources have different pattern profiles. Metabolomic fingerprinting with multivariate data analysis is a reliable and comprehensive technology for authentication and identification. With application of HPTLC imaging,155,211 MS fingerprinting,147,148 or NMR profiling,120–122,212 ginseng saponins,147,148 amino acids,136,212 and polyacetylenes91 can account for these variations.

The total ginsenoside content of P. notoginseng is higher than that of P. quinquefolius and much higher than that of P. ginseng. One difference between the three species is the unique presence of ginsenoside Rf in P. ginseng.213 Ginsenoside Rb2 is detected in abundance in P. ginseng but in trace amounts in the other two species.18 Notoginsenoside R1 is characteristic of P. notoginseng, while pseudoginsenoside F11 is a marker compound in P. quinquefolius. F11 possesses the same molecular weight and has similar retention times as Rf.141 F11 without chromophores cannot be detected by UV; HPLCELSD140 or HPLCMS164 are capable of detecting both F11 and Rf. A UPLC/Q-TOF-MS was developed for 2-min analyses of Rf and F11 in P. ginseng-adulterated P. quinquefolius.164 Establishing the ratios of Rg1/Re, Rg1/Rb1 and Rb2/Rb1 is also useful. Ratios less than 0.4 indicate P. quinquefolius; higher ratios are characteristic of P. ginseng.140,164 One exception is the high Rg1/Rb1 ratio found in wild P. quinquefolius.214 Other spectrometric methods have been reported for effective differentiation, such as 2-D correlation FTIR spectroscopy128 and 2-D NMR spectroscopy.122

4.1.2 Molecular marker. Inspecting genetic makeup is an alternative to defining botanical identity. The genetic makeup of a herbal species does not vary with its physical form or physiological and external conditions. With advances in molecular biotechnology, genetic tools are reliable for authentication of ginseng species at the DNA level. In 2003, Hon et al.215 reviewed genetic methods used in the authentication of Panax species. These genetic tools included arbitrarily-primed PCR,216 random amplified polymorphic DNA (RAPD),217 sequence-characterized amplified regions (SCARs),218 low-Cot DNA fingerprinting,219 restriction fragment length polymorphism (RFLP),217 amplified fragment length polymorphism (AFLP),220 mutant allele specific amplification,221 direct amplification of length polymorphism (DALP) analysis,222 and microsatellite marker technology.12

Zhu et al.223 revealed the phylogenetic relationships between thirteen Panax taxa by DNA sequence analysis on the chloroplast trnK gene and nuclear 18S rRNA gene. Subsequently, they developed a multiplex amplification refractory mutation system for the identification of five commonly used Panax species, i.e., P. ginseng, P. japonicus, P. quinquefolius, P. notoginseng and P. vietnamensis.224 Qin et al.174 developed a microchip electrophoresis method coupled with a PCR–short tandem repeats technique to discriminate between P. quinquefolius and P. ginseng and also between cultivated and wild P. quinquefolius. Diao et al.225 developed a PCR-RFLP and amplification refractory mutation system for identifying P. ginseng from its adulterants. P. ginseng was successfully identified in single herbal medicines or preparations containing diverse components. Sasaki et al.226 developed a loop-mediated isothermal amplification PCR (LAMP) method that could be conducted under isothermal conditions. Lu et al.227 established nested PCR and DNA sequencing methods to identify P. ginseng in preparations. With this method, analytical time and costs were reduced. Table 6 summarizes the advantages and limitations of molecular biology methods.

Table 6 Advantages and limitations of DNA profiling methods for authentication of ginseng species
Methoda Principle Advantages Limitations Ref.
a Abbreviations: RAPD: random amplified polymorphic DNA; SCAR: sequence-characterized amplified region; RFLP: restriction fragment length polymorphism; AFLP: amplified fragment length polymorphism; DALP: direct amplification of length polymorphism; LAMP: loop-mediated isothermal amplification of polymerase chain reaction (PCR).
RAPD Use of small arbitrary primers to produce multiple DNA fragments Technical simplicity, no sequence information required, polymorphism identification Low reproducibility, extra amplified fragments 228–232
 
SCAR Extension of RAPD primers based on polymorphic fragment sequences Reduced competition between primer binding sites and high reproducibility Low sensitivity 218
 
RFLP Comparison of band profiles after restriction enzyme digestion of target DNA. High reliability, no sequence information required Large DNA amounts required, low polymorphism levels 70,217,225,233
 
AFLP Polymorphism based on gain or loss of restriction site, or selective bases High reproducibility, no prior sequence knowledge required Difficulty in identifying homologous markers 220,234,235
 
DALP Use of an arbitrarily primed-PCR to produce genomic fingerprints and to enable sequencing of DNA polymorphisms High resolution fingerprint, new genetic markers characterization Low reproducibility 222
 
DNA sequencing Measurement of every base in the DNA sequence Commonly used, application to primer designation Time-consuming 227
 
Microsatellite Determination of the short tandem repeats at a location Highly polymorphic and codominant High cost and highly time-consuming 220,236,237
 
LAMP A single-tube technique for the amplification of DNA Reduced reaction time, high sensitivity Low specificity 226


4.2 Quality assessment

As global market demands for ginseng increase, quality control has become important. Unlike chemical drugs with a single entity, ginseng, like fruits and vegetables, has many constituents. Natural variations in soil and climate and preparation procedures affect the safety and batch-to-batch uniformity of ginseng products. Monitoring for quality should be done at selected steps for the dynamic coordination of all links. Good ginseng materials and manufacturing processes depend on good agricultural practices (GAPs) and good manufacturing practices (GMPs). Independent quality evaluation is an integral part of ginseng production. To standardize a ginseng extract, metabolomic fingerprinting is performed or marker compound(s) determined.
4.2.1 Plant metabolomic fingerprinting. Metabolomic fingerprinting has been accepted internationally for assessment of herbal medicines. The basic concept of the fingerprint pattern is to consider the whole chromatographic profile or the whole spectrum as a feature.1 The entire pattern of compounds can then be evaluated to determine not only the presence of desired markers but also the complete set of ratios of all detectable analytes.155 Fingerprinting can match the inherent holistic properties of herbs and offers a logical tool for species authentication, quality evaluation, and lot-to-lot consistency.

Table 7 lists some examples of ginseng analysis using the metabolomic fingerprinting technique. The analytical tools include MS-based chromatography and NMR-based spectroscopy.118 Various constituents can be identified and quantified at low concentrations, making MS a sensitive analytical tool for metabolic fingerprinting.136,146,148 Abundant fragment ions by multi-stage MS and high mass accuracy by TOF-MS offer structural information and the possibility of identifying unknown components. In NMR-based fingerprinting, the 1H NMR spectrum provides a wealth of chemical information on ginseng.212,238 NMR has the highest reliability in metabolomics.118 Apart from ginseng saponins, other important metabolites in ginseng such as carbohydrates, amino acids and organic acids can be elucidated based on their chemical shifts in the 1H NMR spectra. The concentration of each metabolite in the sample can be easily calculated from the integration of the signals in the spectra. There is no requirement for calibration curves to convert signal intensity into concentration. When the spectral complexity and signal overlap in the 1H NMR spectra are high, the chemical structures of metabolites can be confirmed by diverse 2D-NMR spectroscopic methods.122

Table 7 Application of metabolomic fingerprinting to analysis of ginseng samples
Instrumenta Chemometricsb Material Result Ref.
a Abbreviations: HPTLC: high-performance thin layer chromatography; 2-D NMR: two-dimensional nuclear magnetic resonance; GCTOF-MS: gas chromatography time-of-flight mass spectrometry; UPLC–ESI-Q-TOF-MS: ultra-performance liquid chromatography electrospray ionization quadrupole-TOF-MS. b Abbreviations: CASE: computer-aided software engineering; PCA: principal component analysis; PLS-DA: partial least squares-discriminant analysis.
HPTLC fluorescent CASE software P. ginseng, P. quinquefolius, P. notoginseng Species authentication and stability of ginseng preparations achieved 155,211
1H-NMR PCA Fresh ginseng roots of different ages Effective authentication and quality control 238
1H-NMR PCA and cluster analysis P. ginseng roots of different origins Sugar signals and methyl signals separate, showing origin-dependent and age-transitions 120,121
1H-NMR PCA and PLS-DA P. ginseng, P. quinquefolius of different origins, and Korean ginseng products Glucose, fumarate, and various amino acids serve as biomarkers for quality assurance 212
2-D J-resolved 1H-NMR PCA Different ages of P. ginseng and commercial ginseng capsules Alanine, arginine, fumaric acid, inositol and ginseng saponins are important metabolites for differentiation 122
GCGCTOF-MS PCA Radix ginseng volatile oils of different ages α-Cadinol, α-bisabolol, thujopsene, and n-hexadecanoic acid increase with age of the sample 136
UPLC–ESI-Q-TOF-MS PCA Different parts of P. notoginseng Slight variations discerned due to different geographical locations, cultivations and collection times 146
UPLC–ESI-Q-TOF-MS PCA and PLS-DA P. ginseng, P. quinquefolius, and P. notoginseng Chikusetsusaponin IVa, Ro, Rc, Rb1, Rb2 and Rg2 account for variations among ginseng species 147
UPLC–ESI-Q-TOF-MS PCA P. ginseng from China and Korea, P. quinquefolius, P. notoginseng, and P. japonicus Rf, F11, malonyl-Rb1, and Rb2 account for variations in species 148


When the data obtained are analyzed by statistical methods, all relevant information must be extracted from a complex data set. Multivariate data analysis is used to recognize patterns and find discriminating signals. Among the multivariate data analysis, PCA146,238 and partial least squares-discriminant analysis (PLS-DA)148,212 have become the routine processing steps for raw analytical data. Marker signals for differentiation can then be identified, and the upper and lower limits of the signals are defined for quality control of ginseng.

4.2.2 Marker compound(s) quantification. Ginseng saponins are selected as the marker compound for the quality control and standardization of ginseng in official monographs. In 1993, the American Botanical Council started the Ginseng Evaluation Program.13 Ginsenosides Rb1, Rb2, Rc, Re, Rg1 and Rd were considered markers for P. ginseng. In 1999, the Word Health Organization published monographs on selected medicinal plants (http://apps.who.int/medicinedocs/en/d/Js2200e/19.html%23Js2200e.19). In that document, ginsenoside Rg1 of not less than 1.5% was selected as the marker compound. In 2002, the European Pharmacopoeia published a monograph on P. ginseng roots that chose ginsenosides Rb1 and Rg1 in amounts not less than 0.4% as markers. In 2004, the American Herbal Pharmacopoeia published monographs for P. quinquefolius and P. ginseng roots and extract. Ginsenosides Rb1, Rb2, Rc, Re, Rg1 and Rd were assigned as the marker compounds. Their total content, as evaluated by HPLC–UV, could not be less than 3% for P. ginseng extracts, 4% for P. quinquefolius roots, and 10% for the extracts. P. ginseng roots could not contain less than 0.2% Rg1 and 0.1% Rb1. In 2009, Health Canada published a monograph on P. ginseng (http://www.hc-sc.gc.ca/dhp-mps/prodnatur/applications/licen-prod/monograph/mono_panax_ginseng-eng.php), which stipulated that ginseng preparations must contain 0.024–0.294 g total ginseng saponins per daily dose.

For a comprehensive evaluation, laboratory methods use as many markers as possible. Kim et al.180 evaluated white and red P. ginseng through simultaneous determination of 14 ginseng saponins by HPLCELSD. Sun et al.17 studied Korean white, red and black P. ginseng by measuring 19 ginseng saponins. Qian et al.239 used HPLCDAD and MS for simultaneous determination of flavonoid, saponins and polyacetylenes in ginseng samples. Comparisons were made of ginseng for the effects of different origins, ages, cultivation methods, plant parts, and commercial manufacturer.3,104,139,214,240,241 Individual and total ginsenoside contents varied in different commercial ginseng products.139 Ginsenoside profiles and content also varied depending on the plant parts tested – in decreasing order of ginsenoside content, they were: leaf > root-hair > rhizome > root > stem.28

To quantify marker compound(s), high-quality ginsenoside libraries are required. Lack of reference standards, in spite of improved methods for determining compounds, makes it difficult to evaluate ginseng products. Some reference compounds are commercially available, but their cost and limited supply cannot meet ginseng demand worldwide. Since most chemical compounds in a herb have a similar skeleton, researchers have introduced a relative quantification correction standard that uses a universal compound for calibration against other structurally similar components. In one study, a commercially available compound, verticinone, was used to quantify twenty-six other steroidal alkaloids.192 Compared with absolute quantitative determinations, an average accuracy of 98.5% was obtained. We have attempted to use Rb1 and Rg1 to determine PPD-type and PPT-type ginseng saponins, respectively, and method validation and robustness tests are still under investigation.

Because the active compounds are unknown, the common practice is to select one or more major ginsenosides such as Rb1 and Rg1 as active or markers. The drawback is less of a link between quality and activity. A combined method of quantifying marker compounds with profiling metabolomic fingerprinting might be a better way to link metabolic profiles with activity and to assure batch-to-batch consistency.

4.2.3 Online structural characterization. A challenge in plant metabolomic fingerprinting is secondary metabolite identification. Traditionally, isolation and NMR-based identification has been the best strategy for absolute determination of structures. However, many constituents are not easily isolated, and most reference compounds are not available in laboratories. The coupling of HPLC and MS has helped on-line structural characterization and rapid identification of compounds in complex herbal extracts.170,206,207,242 The application of tandem MS or, more recently, IT-TOF-MS and Q-TOF-MS, also has advanced the on-line identification of constituents in ginseng samples.186,187,190,243,244

Fig. 7 shows one method for rapid identification of targeted and untargeted ginseng saponins. Available reference compound(s) are used to obtain characteristic fragmentation pathways and diagnostic ions of ginseng saponins. The diagnostic ions and pathways represent the mother nucleus and some substituent groups of saponins. For instance, our ongoing investigations revealed that all PPD-type ginseng saponins produce three common skeleton ions by TOF-MS, i.e., m/z at 407.37, 425.37 and 443.38. Sugar types, numbers and linkage sequences also can be determined by simultaneous or successive losses of sugar moieties and sugar cross-ring cleavages.189


General strategy for identification of ginseng saponins in ginseng samples by mass spectrometry (MSn and accurate mass experiments).
Fig. 7 General strategy for identification of ginseng saponins in ginseng samples by mass spectrometry (MSn and accurate mass experiments).

Following this analysis, the complex ginseng extract is tested. Chromatographic separation by LC is important for subsequent identification. Diagnostic ions and fragmentation mechanisms from reference compounds are a basic requirement for screening known and unknown compounds in ginseng extract. Target compounds can be unequivocally identified by comparison of accurate retention times, molecular ions, and characteristic fragment ions with those of the reference compounds. Identification of untargeted ginseng saponins is complex. The first step is to determine the molecular ions of chromatographic peaks in the ginseng extract. Then, several criteria are used to calculate the accurate molecular formula of each peak: the acceptable accuracy threshold, the general rule of the number of nitrogen atoms, the dibasic ester (DBE) index, and the “show isotopic pattern” function. With the obtained molecular formula, screening for a hit is subsequently performed against various chemical databases. From the candidates collected, the characterized diagnostic ions can then be used for locating the candidates that contain such a substructure and/or substituent groups. Finally, the most likely structure can be determined from these candidates by fragmentation screening and matching.

Online structure characterization has been used to identify 152 saponins from P. notoginseng by LC–ESI-MSn;188 while 35 ginseng saponins in P. ginseng187 and 30 ginseng saponins in P. quinquefolius186 were identified by LCAPCI-MS.

4.2.4 Dynamic chemical transformation. Dynamic structural and concentration changes of ginseng saponins take place during storage, processing, and extraction procedures. Transformation was observed in the class of malonyl ginseng saponins and neutral ginseng saponins (commonly simply referred to as ginseng saponins).

Malonyl ginseng saponins, also called acidic ginseng saponins, are compounds with a malonyl group attached at the 6′′-position of the glucosyl moiety. As early as 1983, four malonyl ginseng saponins, namely the malonyl-ginsenosides Rb1, Rc, Rb2 and Rd, were isolated from P. ginseng245 and later from P. quinquefolius.246 Malonyl-ginsenoside Ra372 and malonyl-notoginsenoside R473 were recently isolated from the fresh roots of P. ginseng. Approximately fifteen malonyl ginseng saponins were found in ginseng extract by HPLCMS/MS.186,187,194,244 Malonyl ginseng saponins, more polar and water-soluble than neutral ginseng saponins, are extremely unstable and susceptible to thermal degradation during drying, storage, heating and extraction.29,247,248 In dried ginseng roots, malonyl ginseng saponins decreased as the air temperature increased.29 Malonyl ginseng saponins are absent (or found in trace amounts) in red ginseng prepared by a steaming or heating process.248 Cold alcohol is recommended to extract malonyl ginseng saponins, and 40% ethanol has maximum extraction efficiency.29 Malonyl ginseng saponins are thus not often found in commercial products prepared by hot extraction processes.190

Malonyl-ginseng saponins make up much of the total saponins in ginseng. Their concentration accounts for approximately one-third of the total ginsenoside content in P. quinquefolius, but relatively little in P. ginseng and P. notoginseng.249,250 Malonyl-ginsenoside Rb1 is the most abundant.11 The profiles of malonyl-ginseng saponins differ among different species.194,249 Because of their instability and the lack of suitable reference compounds, determining their profiles presents a challenge. Since they are usually invisible on HPLC–UV chromatograms, standard assay procedures may underestimate ginsenoside content by ignoring malonyl ginsenosides. Use of phosphate buffer is important for resolution of malonylated and other esterified ginseng saponins.247,251 If ELSD or MS is used for detection, ammonium acetate,244 ammonium formate,190 and acetic acid194 are recommended instead of phosphate. An alternative strategy is to determine malonyl ginseng saponins by an indirect two-step method that hydrolyzes these acidic saponins into their respective neutral saponins with KOH. However, the results may give only a rough estimate because other derivatives also might be transformed. Both APCI-MS186,187 and ESI-MS16,194,244 are useful for characterization of malonyl-ginsenosides in ginseng extracts. Fig. 8 shows the MS characterization of malonyl-ginsenoside Rb1 by tandem MS in negative ion mode. Fig. 8A lists the sequential mass spectra of deprotonated malonyl-ginsenoside Rb1, and Fig. 8B suggests the fragmentation pathways. The presence of a free carboxylic acid group means that malonyl ginseng saponins can be rapidly identified as they display loss of the neutral units of mass 44 Da (CO2), 42 Da (CH2[double bond, length as m-dash]CO) and 60 Da (CH3COOH).


Mass spectrometry (MS) characterization of malonyl-ginsenoside Rb1 by tandem MS in negative ion mode. (A) Sequential mass spectrometric analysis of deprotonated malonyl-ginsenoside Rb1. (B) Interpretation of the fragmentation pattern. Experimental conditions are given in ref. 194.
Fig. 8 Mass spectrometry (MS) characterization of malonyl-ginsenoside Rb1 by tandem MS in negative ion mode. (A) Sequential mass spectrometric analysis of deprotonated malonyl-ginsenoside Rb1. (B) Interpretation of the fragmentation pattern. Experimental conditions are given in ref. 194.

Neutral ginseng saponins, usually stable during drying and storage conditions,252 degrade to new compounds under intensive heating or steaming. The conversion means that ginsenoside profiles of unprocessed and processed ginseng are different.52,55,253,254 As shown in Fig. 9, during steaming or heating, the polar ginseng saponins decreased, and less polar ginseng saponins increased. The structural changes in ginsenosides during the process are elimination of sugar chains and isomerization of the hydroxyl configuration at C-20 of the aglycones.52 The protopanaxadiol (PPD)-type ginsenosides selectively eliminate the C-20 sugar chain to produce Rg3.255,256 Rg3 is further transformed to Rk1 and Rg5 by dehydration.3 A small amount of Rh2 observed in red ginseng implies that the elimination of C-3 sugar residue is relatively difficult in processing.55 The protopanaxatriol (PPT)-type ginsenosides first lose the C-20 sugar residue and subsequently their terminal sugar unit at C-6 to form Rg2 and/or Rh1.248 Rh1 is further converted to Rk3 and Rh4 by dehydration at C-20. In summary, the elimination of sugar chains at C-20, then at C-6 or at C-3, and a subsequent dehydration reaction at C-20 are the effects of steaming.


HPLC–UV chromatograms of ginseng roots and chemical degradation in steaming process, modified from ref. 256. (A) Ethanol extract of roots of P. quinquefolius, P. ginseng and P. notoginseng. (B) Ethanol extract of steamed ginseng roots at 120 °C for 4 h. Chromatographic separation was performed on a Prodigy ODS column. Detection wavelength was set at 202 nm. The mobile phase used acetonitrile and water for a gradient elution.
Fig. 9 HPLC–UV chromatograms of ginseng roots and chemical degradation in steaming process, modified from ref. 256. (A) Ethanol extract of roots of P. quinquefolius, P. ginseng and P. notoginseng. (B) Ethanol extract of steamed ginseng roots at 120 °C for 4 h. Chromatographic separation was performed on a Prodigy ODS column. Detection wavelength was set at 202 nm. The mobile phase used acetonitrile and water for a gradient elution.
4.2.5 Pesticide determination. Pesticide and heavy-metal contamination are safety concerns with ginseng products. Ginseng roots require 3–6 years of cultivation until harvest. During this time, disease control with pesticides against insects and environmental pollution means that the ginseng plants accumulate chemical contaminants. Multiple pesticides such as organochlorine,257 organophosphorus,193 organohalogen,160 and pyrethroid components258 are found to be above the limits of detection, or even over maximum residue limits, in ginseng products. Few studies have been devoted to investigating contamination of ginseng with pesticides or toxic metals.

The steps in pesticide analysis are extraction, cleanup, separation and detection.259 Ginseng samples are blended with organic solvent alone, solvent mixtures, or water. Acetone,159,260 acetonitrile,159,261 methanol,262 and ethyl acetate160 are used. Pesticide extraction methods are Soxhlet extraction,257,263 solid-phase extraction (SPE),159,261 matrix solid-phase dispersion,61,196 and SFE.264,265 Traditional cleanup procedures fractionate extracts based on polarity, as in liquid–liquid partitioning257,258 and column chromatography, or adsorption chromatography using florisil196,260,266,267 and neutral alumina.260,268 SPE265,268 and gel permeation chromatography160 have become popular. GC is used for separation of large-scale multiresidue pesticides.159,160,266 Because some pesticides are highly polar, have low volatility and/or are thermo-labile, LC is used for analysis.61 Element-selective electron capture,258,261,263,267 nitrogen-phosphorus analysis,258,261 and flame photometrics are used.193 The detectors are sometimes connected to combine results in a single run.261,263,267 MSn and TOF-MS are the most commonly used detection techniques.159,160,267,268

In a survey of P. ginseng and P. quinquefolius extracts by Durgnat et al.,269 13 out of 30 samples contained levels of pesticides greater than the maximum residue limits. When Hayward et al.159,160 analyzed 170 organohalogen and organophosphorous pesticides and their isomers and metabolites in dried ginseng roots, seven products contained combinations of pesticides.

4.3 Interaction with biological systems

4.3.1 Pharmacokinetic parameters. Pharmacokinetic (PK) evaluation of ginseng is challenging because of the diversity of ginseng saponins. Table 8 summarizes PK investigations into ginseng and ginseng saponins. The major parameters, Tmax, T1/2 and F (bioavailability), are included.
Table 8 Pharmacokinetic data on ginseng and ginseng saponins
Materiala Administrationb (species) Analytical method, LLOQ c (ng ml−1) T max (h) T 1/2 (h) F (%) Ref.
a PPD: 20(S)-protopanaxadiol; IH-901 (or compound K): 20-O-(β-D-glucopyranosyl)-20(S)-protopanaxadiol; “Shenmai”: a prescription prepared from Radix Ginseng and Radix Ophiopogonis. b i.v.: intravenous; i.g.: intragastrical; i.m.: intramuscular. c LLOQ: lower limit of quantification; UPLC: ultra-performance liquid chromatography; ESI-MS: electrospray ionization-mass spectrometry; APCI: atmospheric pressure chemical ionization; Q-TOF: quadrupole time-of-fight; SSIM: segmental and selected ion monitoring. d T max: time to reach the maximum plasma concentration after i.g. or oral administration. e T 1/2: elimination half-life. f F: oral bioavailability; ND: not detected in plasma after oral administration.
P. ginseng berry and Re i.v. and i.g. (rat) UPLCESI-MS, 2.5 0.4 ± 0.2 0.2–0.5 0.19–0.28 270
Rd i.g. and i.v. (dog) LCESI-MS, 5 3.0 24.2 ± 2.85 for i.g., 39.4 ± 12.0 for i.v. 0.26 271
Rd i.v. (human) LC–ESI-MS-MS, 3 0.5 ± 1.0 19.3 ± 3.4 272,273
Rg1 i.v. (rat) LCESI-MS, 2–10 1.33 274
Rh1 i.g. and i.v. (rat) LCESI-MS, 5 1.00 ± 0.00 0.43 ± 0.08 for i.g., 0.41 ± 0.05 for i.v. 1.01 ± 0.03 275
20(S)- and 20(R)-Rg2 i.v. (rat) LC–UV, 3900–7800 0.64 ± 0.02 for 20S, 1.19 ± 0.06 for 20R 276
20(S)-Rg2 i.g. and i.v. (rat) LCESI-MS, 5 0.53 ± 0.34 ND 277
20(S)-Rg3 i.g. and i.v. (rat) LCESI-MS, 2–10 0.23 2.63 278,279
20(S)-Rg3 i.m. (human) LCESI-MS/MS, 2.0–2.5 280
20(R)-Rg3 i.g. and i.v. (rat) LCESI-MS, 30 0.31 for i.v. ND 281
20(R)-Rh2 i.g. and i.v. (rat) LC–ESI-Q-TOF-MS 0.27 ND 282
IH-901 i.g. and i.v. (rat) LCESI-MS/MS, 5 0.63 ± 0.18 3.70 ± 1.38 for i.v. 4.54 ± 0.38 283,284
PPD i.g. and i.v. (rat) LCAPCI-MS, 1 2.50 ± 1.23 1.47 ± 0.30 for i.g., 2.73 ± 0.85 for i.v. 36.8 ± 12.4 285
PPD oral (human) LCESI-MS/MS, 10 1.28 ± 0.49 4.77 ± 2.05 286
25-OH-PPD i.g. and i.v. (rat) LCESI-MS/MS, 10 5.50 ± 4.70 3.9 ± 2.1 for i.g., 4.5 ± 2.6 for i.v. 64.8 ± 14.3 287
P. notoginseng extract i.g. and i.v. (rat) LC–UV, 1200–4000 18.57 for Rb1, 14.13 for Rg1 4.35 for Rb1, 18.4 for Rg1 288
P. notoginseng extract i.g. and i.v. (rat) LCESI-MS, 2.8–4.0 0.71 ± 0.19 for R1, 0.75 ± 0.00 for Rg1, 0.88 ± 0.14 for Rd, 0.79 ± 0.10 for Re, 0.83 ± 0.13 for Rb1 1.11 ± 0.51 for R1, 5.01 ± 2.09 for Rg1, 18.15 ± 9.14 for Rd, 1.01 ± 1.35 for Re, 20.15 ± 6.27 for Rb1 9.29 for R1, 6.06 for Rg1, 2.36 for Rd, 7.06 for Re, 1.18 for Rb1 289
Ginseng saponins i.g. (rat) LCESI-MS, 8–50 290
Shenmai i.v. (human) LC–ESI-MS-MS, 1 2.09 ± 1.89 for Rg1 291
Shenmai i.v. (rat) UPLC–SSIM-MS, 1–10 292


Sample pretreatment and analysis is key in PK studies. A simple sample pretreatment uses protein precipitation and liquid–liquid extraction. Another sample pretreatment method uses SPE to remove interference and matrix effects. SPE suffers from lower extraction recoveries than liquid–liquid extraction. For analysis, LC–UV and LCMS have been developed to assay ginseng saponins in biological samples. LC–UV is convenient but has low sensitivity with a lower limit of quantification (LLOQ) greater than 1000 ng ml−1. LCMS and MS-MS are more sensitive (LLOQ, 1–10 ng ml−1) and specific. Segmental and SIM have better sensitivity and wider dynamic ranges than SIM alone.292 PPD and PPT aglycone have weak ionization efficiency in MS compared with saponins, which possess sugar moieties.290 Digoxin is generally selected as the internal standard for MS quantification.

The oral bioavailability of PPD-type saponins (ginsenosides Ra3,293 Rb1,289 Rd,271 Rg3,278 and Rh2294) and PPT-type saponins (ginsenosides Rg1,274 Re,270 Rh1,275 and notoginsenoside R1289) is less than 5%.295 Excretion of ginseng saponins at 0.2–1.2% can be expected in human urine.296 Extensive metabolism in the gastrointestinal tract,279,297 poor membrane permeability,293 and lowered solubility of deglycosylated products298 limit intestinal absorption of ginseng saponins. Higher oral doses might saturate metabolism and increase the bioavailability.284,288 PPT-type saponins have better bioavailability than that of PPD group,288,289 perhaps because PPD-type saponins degrade faster than the PPT-type. IH-901 or compound K, an intestinal bacterial metabolite of PPD-type saponins, contributes to low bioavailability because of biliary excretion and hepatic metabolism via esterification with fatty acids.283,284 Changing the pharmaceutical formulation may improve bioavailability. For instance, micronized Rh2 has a doubled bioavailability;298 the inclusion complex of IH901 and the water-soluble agent β-cyclodextrin offers 1.9-fold higher bioavailability than pure IH901 powder.299 The bioavailability of PPD aglycone and 25-OH-PPD is improved because of less metabolism and favorable membrane permeability.285–287

The time to reach maximum concentration (Tmax) in plasma and tissues is generally less than 2 h, indicating that ginseng saponins are rapidly absorbed and readily distributed in the tissues.276,300 Levels of a ginseng saponin in the system depend on its elimination half-life (T1/2). Most ginseng saponins and their deglycosylated products were excreted by the biliary system through active transport, which affected their T1/2 values and systemic exposure.293 Time curves of ginseng saponins exhibited distinct multiple peaks after oral administration, indicating the involvement of enterohepatic recirculation.284

Tissue disposition demonstrated that liver and bile are responsible for systemic clearance of ginseng saponins from the circulation.283,284,300 Hepatic cytochrome P450 catalyzed ginsenoside metabolism has been described, and CYP3A4-catalyzed oxygenation metabolism played an important role in the hepatic disposition of ginsenosides. Table 8 shows that Ra3, Rb1, Rc and Rd have significantly longer T1/2 values (18.57–39.4 h) than other ginseng saponins (0.2–4.77 h). Attachment of more sugar moieties in the PPD-type ginsenosides Ra3, Rb1, Rc and Rd blocks their access to biliary transporters and slows biliary excretion.293 Some PK parameters shown in Table 8 are far from the mean values, such as Tmax and T1/2 of 25-OH-PPD and Rd. Whether this variation is caused by different factors or experimental conditions requires further evaluation.

4.3.2 Metabolic pathways. The metabolic pathways of PPD-type and PPT-type ginseng saponins on animals and humans are proposed in Fig. 10A and Fig. 10B, respectively. Identification of ginsenoside metabolites is generally based on MSn experiments (LC–Q-IT-MS) and/or confirmation by accurate mass measurements (LCQ-TOF-MS). Comparison of the MSn fragment ions of the parent compounds with those obtained from the metabolites provides valuable clues to the parts of the molecule that undergo modification by degradation or combination or that remain unchanged. Sometimes, derivatization of functional groups or isolation of the metabolites from biofluids for further NMR assays can be performed.
Proposed in vivo metabolic pathways of ginseng saponins. (A) protopanaxadiol-type saponins. (B) protopanaxatriol-type saponins. “” denotes major pathways; “” denotes additional pathways. [P]: metabolites detected in plasma; [U]: metabolites in urine; [F]: metabolites in feces; [B]: metabolites in bile. ig: metabolites after oral administration; iv: metabolites after intravenous injection.
Fig. 10 Proposed in vivo metabolic pathways of ginseng saponins. (A) protopanaxadiol-type saponins. (B) protopanaxatriol-type saponins. “ugraphic, filename = c0np00057d-u1.gif” denotes major pathways; “ugraphic, filename = c0np00057d-u2.gif” denotes additional pathways. [P]: metabolites detected in plasma; [U]: metabolites in urine; [F]: metabolites in feces; [B]: metabolites in bile. ig: metabolites after oral administration; iv: metabolites after intravenous injection.

The conversion of ginseng saponins in the gastrointestinal tract also has been studied using in vitro models such as acid conditions, enzymes, intestinal bacteria and microsomes.198,301 Because of high ginsenoside concentration used in the in vitro studies, the metabolic profiles may differ from in vivo results.302 The main metabolic pathways in vivo include deglycosylation reactions in ginseng saponins by intestinal bacteria via stepwise cleavage of the sugar moieties.293,295,297 Oxygenation by intestinal and hepatic CYP450 enzymes is another metabolic pathway.275,281,303 Oxygenation generally occurred on the ‘top-right’ aliphatic chain.282 Parental metabolites that could be further esterified with fatty acids in liver and tissues sustain fatty acid conjugates longer in the body.304 After oral administration of P. notoginseng extract to rats, the most abundant metabolites in rat feces were IH-901, Rg1 and F1.293 PPD and PPT aglycones were detected in low quantities in feces and not detected in plasma, bile and urine.293 After human volunteers ingested Ginsana G115 capsules (4% ginseng saponins), IH-901, Rh1 and F1 were the major metabolites found in the systemic circulation and excreted in urine.297 In some publications, 20(S)- and 20(R)-ginsenosides were not differentiated.278–281 Only a few studies have compared the metabolism and pharmacokinetics of 20(S)- and 20(R)-ginsenosides.276

Because of competitive absorption and metabolism between the various components, differences can be observed between the administration of a single ginsenoside and of ginseng extract. The metabolic profile of a ginsenoside can also vary with different methods of administration. For instance, Rb1 is the dominant metabolite after intravenous injection of Rd, and Rg3 is the major metabolite after oral administration of Rd.273

4.3.3 Endogenous metabonomics. Metabonomics is the comprehensive systematic study of metabolite levels and changes in organisms over time as a consequence of stimuli such as disease or pharmaceutical interventions.305 Metabonomics has become a tool to evaluate the curative effects and the safety of herbal medicines including ginseng.138,306–308 Through metabonomic analysis, biomarkers are recognized and the basic dynamic variations of metabolic responses to herbal medicine intervention are revealed. With such information, the knowledge of a disease may improve, onset of disease may be predicted, and the clinical efficacy, safety, and mechanisms of action of complex herbal medicines can be evaluated.

The principal analytical techniques for metabonomic studies are based on NMR spectroscopy and mass spectrometry. All metabonomic studies result in complex, multivariate datasets that require visualization software and chemometric and bioinformatic methods for interpretation. Wang et al.138 reported metabolic regulatory network alterations in response to acute cold stress in the rat and changes after ginsenoside intervention. Urinary metabolite profiling using GCMS with multivariate statistical techniques revealed biochemical changes of these metabolites and demonstrated the protective effect of total ginseng saponins. Several metabolic pathways were identified in metabolic regulation and compensation to restore homeostasis. Wang et al.309 performed a metabonomics study on the effects of the ginsenoside Rg3 on liver-tumor-bearing rats. They combined column-switching with the orthogonal selectivity of hydrophilic interaction chromatography and reversed-phase chromatography. The administration of a single, high dose of Rg3 changed the metabolic pattern in cancer rats during the three days studied, and 17 biomarker candidates were identified.

For such studies, complex data must be interpreted and combined with analytical methods. One problem is that the number of metabolites produced by a given system is difficult to predict. Given the potential interest in this field, metabonomics might be useful for a comprehensive evaluation of the systemic clinical efficacy, safety, and mechanism of action of ginseng.

4.3.4 Interaction with biomacromolecules. The interaction between small molecules (e.g., ginsenosides) and biomacromolecules (e.g., proteins and DNA) is one paradigm in pharmaceutical research.310 To discover target candidates for new drugs, the mechanisms and binding parameters (e.g., dissociation constants) between ginseng constituents and biomacromolecules should be characterized. Various strategies have emerged for the systematic and high-speed characterization of the interaction of small molecules with a biomacromolecule at the molecular level.310 Most of the techniques study the simple binding system in which one constituent of ginseng binds to one biomacromolecular receptor.311,312 There is a trend towards library screening (e.g., ginseng extract) or identifying interacting counterparts against known or unknown libraries.310,313

Zou et al.314,315 proposed biological fingerprinting to investigate the binding characteristics of natural products with target biomacromolecules that are free or immobilized in affinity chromatography. Bioactive compounds that specifically bind target biomacromolecules can be screened from complex herbal medicines. Li et al.1 developed holistic methods and integrated evaluation models to investigate biomacromolecule-binding compounds in ginseng preparations. Crude extracts are divided into two major fractions, namely, fractions unrelated to bioactivities and fractions related to bioactivities. The latter fraction is then characterized by on-line HPLCMS. Structural information and binding parameters of each captured compound can be determined. A good example is the recent development of 2-D turbulent flow chromatography (TFC) coupled on-line to LCMS for solution-based ligand screening against multiple proteins, shown in Fig. 11.313 This technique is an efficient tool for high-throughput screening of library ligands with low-to-high binding affinities and for evaluating the structure–activity relationship.


Schematic diagram of the two-dimensional turbulent flow column-switching LC–MS apparatus, modified from ref. 313. Ginseng extract or ginseng saponin is incubated with the target biomacromolecule. The first turbulent flow chromatography (TFC), which only retains low-molecular-weight analytes, is used to separate free compounds and the protein–ginsenoside complex. The captured complex is broken up in the reaction wire by a dissociation solution. The compounds released are separated and enriched from biomacromolecules by the second TFC. On-line LC–MS is used to determine the structural information and affinity parameters of the binding compounds.
Fig. 11 Schematic diagram of the two-dimensional turbulent flow column-switching LCMS apparatus, modified from ref. 313. Ginseng extract or ginseng saponin is incubated with the target biomacromolecule. The first turbulent flow chromatography (TFC), which only retains low-molecular-weight analytes, is used to separate free compounds and the protein–ginsenoside complex. The captured complex is broken up in the reaction wire by a dissociation solution. The compounds released are separated and enriched from biomacromolecules by the second TFC. On-line LCMS is used to determine the structural information and affinity parameters of the binding compounds.

Wang et al.316 screened anti-platelet aggregation agents from P. notoginseng using human platelet extraction and HPLC–DAD-ESI-MS/MS analysis. Five compounds that bind with human platelets were detected, and four were identified as adenosine, guanosine, ginsenoside Rh1 and F1. Liu et al.317 studied the interaction of total saponins of P. notoginseng with human serum albumin (HSA) by FTIR spectroscopy. Ginseng saponins were shown to combine with HSA through C[double bond, length as m-dash]O and C[double bond, length as m-dash]N groups of polypeptide chain. The ginsenoside–protein combination caused a significant loss of α-helix structure and changes of the tyrosine residues in protein. Dasgupta et al.318 compared the effects of five different ginseng species on serum digoxin measurement by immunoassays. Digibind, an antidigoxin antibody, can bind free digoxin-like immunoreactive components of ginseng. Thus, the ginsenoside–biomacromolecule interaction reveals complex interconnected biological networks, which may help with discovery of new protein targets and drug candidates.

5 Concluding remarks and perspectives

Compounds isolated from ginseng are an abundant source of chemical diversity from which to discover active molecules. Synthesis or biosynthesis is the preferred way to produce novel compounds for pharmacological activities. Synthesis also helps us understand structure–activity relationships. Besides sample preparation and purification steps, current isolation protocols often comprise in vitro assays, which are frequently coupled on-line to HPLC or MS systems.

A major challenge of ginseng research is the complexity of chemical constituents, especially polar macromolecules. However, technical advances in analytical tools are promising. Chromatography shows a trend towards faster and more comprehensive separations, indicated by wide applications of UPLC, HILIC and multidimensional chromatography. Recent innovations have made mass spectrometers more sensitive and selective, and MS and tandem MS are now applied in numerous analytical fields of ginseng research.

One approach to investigating ginseng is to view the extract as an indivisible whole, a self-organized and interactive multicomponent entity. Plant metabolomic fingerprinting and metabonomics of ginseng-treated biological samples are holistic approaches to quality control and activity evaluation, respectively. Other better integral analyses are needed to measure complex ginseng constituents, their metabolites and other related macromolecules.

Ginseng analysis requires expertise in chemistry, chemometrics, biology, and bioinformatics. With the exponentially increasing amount of data from ginseng plant and ginseng-treated biological samples, collaboration among chemists, biologists, and computer scientists is important. We believe that there are good prospects for obtaining new insights into drug discovery and clinical utility by the continued study of ginseng.

6 Acknowledgements

Special thanks go to Dr Lorena Tomas Laudo for encouraging us to prepare this review. We also thank Sally Kozlik for editing the manuscript. This work was supported in part by the NIH grants P01 AT004418 and K01 AT005362.

7 References

  1. P. Li, L. W. Qi, E. H. Liu, J. L. Zhou and X. D. Wen, TrAC, Trends Anal. Chem., 2008, 27, 66–77 CrossRef CAS .
  2. L. Jia and Y. Q. Zhao, Curr. Med. Chem., 2009, 16, 2475–2484 CrossRef CAS .
  3. C. Z. Wang, H. H. Aung, M. Ni, J. A. Wu, R. B. Tong, S. Wicks, T. C. He and C. S. Yuan, Planta Med., 2007, 73, 669–674 CrossRef CAS .
  4. A. B. Fishbein, C. Z. Wang, X. L. Li, S. R. Mehendale, S. Sun, H. H. Aung and C. S. Yuan, Arch. Pharmacal Res., 2009, 32, 505–513 Search PubMed .
  5. G. Y. Li, Y. M. Zeng, H. Meng, X. Li and J. H. Wang, Chin. Chem. Lett., 2009, 20, 1207–1210 CrossRef CAS .
  6. J. T. Xie, C. Z. Wang, B. Zhang, S. R. Mehendale, X. L. Li, S. Sun, A. H. Han, W. Du, T. C. He and C. S. Yuan, Biol. Pharm. Bull., 2009, 32, 1552–1558 CrossRef CAS .
  7. J. T. Xie, S. R. Mehendale, A. B. Wang, A. H. Han, J. A. Wu, J. Osinski and C. S. Yuan, Pharmacol. Res., 2004, 49, 113–117 CrossRef CAS .
  8. T. K. Yun, Lancet Oncol., 2001, 2, 49–55 CrossRef CAS .
  9. L. W. Qi, C. Z. Wang and C. S. Yuan, Biochem. Pharmacol., 2010, 80, 947–954 CrossRef CAS .
  10. L. Jia, Y. Zhao and X. J. Liang, Curr. Med. Chem., 2009, 16, 2924–2942 CrossRef CAS .
  11. L. P. Christensen, Adv. Food Nutr. Res., 2009, 55, 1–99 Search PubMed .
  12. N. Angelova, H. W. Kong, R. Van Der Heijden, S. Y. Yang, Y. H. Choi, H. K. Kim, M. Wang, T. Hankemeier, J. Van Der Greef, G. Xu and R. Verpoorte, Phytochem. Anal., 2008, 19, 2–16 CrossRef CAS .
  13. N. Fuzzati, J. Chromatogr., B, 2004, 812, 119–133 CrossRef CAS .
  14. S. J. Kim, H. N. Murthy, E. J. Hahn, H. L. Lee and K. Y. Paek, Sep. Purif. Technol., 2007, 56, 401–406 CrossRef CAS .
  15. J. A. Wood, M. A. Bernards, W. K. Wan and P. A. Charpentier, J. Supercrit. Fluids, 2006, 39, 40–47 CrossRef CAS .
  16. T. Ligor, A. Ludwiczuk, T. Wolski and B. Buszewski, Anal. Bioanal. Chem., 2005, 383, 1098–1105 CrossRef CAS .
  17. B. S. Sun, L. J. Gu, Z. M. Fang, C. Y. Wang, Z. Wang, M. R. Lee, Z. Li, J. J. Li and C. K. Sung, J. Pharm. Biomed. Anal., 2009, 50, 15–22 CrossRef CAS .
  18. J. B. Wan, S. P. Li, J. M. Chen and Y. T. Wang, J. Sep. Sci., 2007, 30, 825–832 CrossRef CAS .
  19. J. H. Kwon, J. M. Belanger and J. R. Pare, J. Agric. Food Chem., 2003, 51, 1807–1810 CrossRef CAS .
  20. Y. T. Wang, J. Y. You, Y. Yu, C. F. Qu, H. R. Zhang, L. Ding, H. Q. Zhang and X. W. Li, Food Chem., 2008, 110, 161–167 CrossRef CAS .
  21. A. S. Engelberth, E. C. Clausen and D. J. Carrier, Sep. Purif. Technol., 2010, 72, 1–6 CrossRef CAS .
  22. J. Wu, L. Lin and F. T. Chau, Ultrason. Sonochem., 2001, 8, 347–352 CrossRef CAS .
  23. Y. Yang, L. Chen, X. X. Zhang and Z. K. Guo, J. Liq. Chromatogr. Relat. Technol., 2004, 27, 3203–3211 CrossRef CAS .
  24. T. T. Dong, K. J. Zhao, W. Z. Huang, K. W. Leung and K. W. Tsim, Phytother. Res., 2005, 19, 684–688 CrossRef CAS .
  25. R. M. Corbit, J. F. S. Ferreira, S. D. Ebbs and L. L. Murphy, J. Agric. Food Chem., 2005, 53, 9867–9873 CrossRef CAS .
  26. S. Gafner, C. Bergeron, M. M. McCollom, L. M. Cooper, K. L. McPhail, W. H. Gerwick and C. K. Angerhofer, J. Agric. Food Chem., 2004, 52, 1546–1550 CrossRef CAS .
  27. M. P. K. Choi, K. K. C. Chan, H. W. Leung and C. W. Huie, J. Chromatogr., A, 2003, 983, 153–162 CrossRef CAS .
  28. C. L. Qu, Y. P. Bai, X. Q. Jin, Y. T. Wang, K. Zhang, J. Y. You and H. Q. Zhang, Food Chem., 2009, 115, 340–346 CrossRef CAS .
  29. X. W. Du, R. B. H. Wills and D. L. Stuart, Food Chem., 2004, 86, 155–159 CrossRef CAS .
  30. H. T. Nguyen, G. Y. Song, J. A. Kim, J. H. Hyun, H. K. Kang and Y. H. Kim, Bioorg. Med. Chem. Lett., 20, pp. 309–314 Search PubMed.
  31. N. H. Tung, G. Y. Song, N. X. Nhiem, Y. Ding, B. H. Tai, L. G. Jin, C. M. Lim, J. W. Hyun, C. J. Park, H. K. Kang and Y. H. Kim, J. Agric. Food Chem., 58, pp. 868–874 Search PubMed.
  32. D. Q. Dou, Y. J. Chen, L. H. Liang, F. G. Pang, N. Shimizu and T. Takeda, Chem. Pharm. Bull., 2001, 49, 442–446 CrossRef CAS .
  33. P. Y. Liao, D. Wang, Y. J. Zhang and C. R. Yang, J. Agric. Food Chem., 2008, 56, 1751–1756 CrossRef CAS .
  34. N. H. Tung, K. Cho, J. A. Kim, G. Y. Song and Y. H. Kim, Bull. Korean Chem. Soc., 2010, 31, 1381–1384 CrossRef CAS .
  35. S. Nakamura, S. Sugimoto, H. Matsuda and M. Yoshikawa, Chem. Pharm. Bull., 2007, 55, 1342–1348 CrossRef CAS .
  36. I. H. Park, N. Y. Kim, S. B. Han, J. M. Kim, S. W. Kwon, H. J. Kim, M. K. Park and J. H. Park, Arch. Pharmacal Res., 2002, 25, 428–432 Search PubMed .
  37. O. Sticher, Nat. Prod. Rep., 2008, 25, 517–554 RSC .
  38. Q. Z. Du, G. Jerz, R. Waibel and P. Winterhalter, J. Chromatogr., A, 2003, 1008, 173–180 CrossRef CAS .
  39. X. C. Qi, S. Ignatova, G. A. Luo, Q. L. Liang, F. W. Jun, Y. M. Wang and I. Sutherland, J. Chromatogr., A, 2010, 1217, 1995–2001 CrossRef CAS .
  40. X. L. Cao, Y. Tian, T. Y. Zhang, Q. H. Liu, L. J. Jia and Y. Ito, J. Liq. Chromatogr. Relat. Technol., 2003, 26, 1579–1591 CrossRef CAS .
  41. Y. W. Ha, S. S. Lim, I. J. Ha, Y. C. Na, J. J. Seo, H. Shin, S. H. Son and Y. S. Kim, J. Chromatogr., A, 2007, 1151, 37–44 CrossRef CAS .
  42. Y. J. Cheng, Q. L. Liang, P. Hu, Y. M. Wang, F. W. Jun and G. A. Luo, Sep. Purif. Technol., 2010, 73, 397–402 CrossRef CAS .
  43. J. Wang, H. L. Bai, C. M. Liu and L. Li, Chromatographia, 2010, 71, 267–271 CrossRef CAS .
  44. Y. Ito, J. Chromatogr., A, 2005, 1065, 145–168 CrossRef CAS .
  45. H. P. Jiang, Y. K. Qiu, D. R. Cheng, T. G. Kang and D. Q. Dou, Magn. Reson. Chem., 2008, 46, 786–790 CrossRef CAS .
  46. J. Chen, R. Zhao, Y. M. Zeng, H. Meng, W. J. Zuo, X. Li and J. H. Wang, J. Asian Nat. Prod. Res., 2009, 11, 195–201 CrossRef CAS .
  47. S. Nakamura, S. Sugimoto, H. Matsuda and M. Yoshikawa, Heterocycles, 2007, 71, 577–588 CrossRef CAS .
  48. W. Wang, Y. Q. Zhao, E. R. Rayburn, D. L. Hill, H. Wang and R. W. Zhang, Cancer Chemother. Pharmacol., 2007, 59, 589–601 CrossRef CAS .
  49. J. Y. Wang, X. G. Li, Y. N. Zheng and X. W. Yang, J. Asian Nat. Prod. Res., 2004, 6, 289–293 CrossRef CAS .
  50. C. Z. Wang, J. A. Wu, E. McEntee and C. S. Yuan, J. Agric. Food Chem., 2006, 54, 2261–2266 CrossRef CAS .
  51. S. Sugimoto, S. Nakamura, H. Matsuda, N. Kitagawa and M. Yoshikawa, Chem. Pharm. Bull., 2009, 57, 283–287 CrossRef CAS .
  52. S. Sun, C. Z. Wang, R. Tong, X. L. Li, A. Fishbein, Q. Wang, T. C. He, W. Du and C. S. Yuan, Food Chem., 2010, 118, 307–314 CrossRef CAS .
  53. S. M. Lee, H. J. Shon, C. S. Choi, T. M. Hung, B. S. Min and K. Bae, Chem. Pharm. Bull., 2009, 57, 92–94 CrossRef CAS .
  54. H. T. Nguyen, G. Y. Song, H. K. Kang and Y. H. Kim, Bull. Korean Chem. Soc., 2010, 31, 2094–2096 CrossRef .
  55. C. Z. Wang, B. Zhang, W. X. Song, A. B. Wang, M. Ni, X. J. Luo, H. H. Aung, J. T. Xie, R. Tong, T. C. He and C. S. Yuan, J. Agric. Food Chem., 2006, 54, 9936–9942 CrossRef CAS .
  56. M. Yoshikawa, T. Murakami, K. Yashiro, J. Yamahara, H. Matsuda, R. Saijoh and O. Tanaka, Chem. Pharm. Bull., 1998, 46, 647–654 CAS .
  57. M. Yoshikawa, S. Sugimoto, S. Nakamura, H. Sakumae and H. Matsuda, Chem. Pharm. Bull., 2007, 55, 1034–1038 CrossRef CAS .
  58. Q. Le Tran, I. K. Adnyana, Y. Tezuka, T. Nagaoka, Q. K. Tran and S. Kadota, J. Nat. Prod., 2001, 64, 456–461 CrossRef CAS .
  59. X. Y. Wang, D. Wang, X. X. Ma, Y. J. Zhang and C. R. Yang, Helv. Chim. Acta, 2008, 91, 60–66 CrossRef CAS .
  60. M. Yoshikawa, T. Morikawa, K. Yashiro, T. Murakami and H. Matsuda, Chem. Pharm. Bull., 2001, 49, 1452–1456 CrossRef CAS .
  61. X. M. Cui, Z. Y. Jiang, J. Zeng, J. M. Zhou, J. J. Chen, X. M. Zhang, L. S. Xu and Q. Wang, J. Asian Nat. Prod. Res., 2008, 10, 845–849 CrossRef CAS .
  62. R. W. Teng, H. Z. Li, D. Z. Wang and C. R. Yang, Helv. Chim. Acta, 2004, 87, 1270–1278 CrossRef CAS .
  63. H. Sun, Y. Ye and Y. Pan, Chem. Biodiversity, 2005, 2, 510–515 CrossRef CAS .
  64. K. Zou, S. Zhu, C. Tohda, S. Q. Cai and K. Komatsu, J. Nat. Prod., 2002, 65, 346–351 CrossRef CAS .
  65. J. M. Jia, Z. Q. Wang and L. J. Wu, Chin. Chem. Lett., 2008, 19, 1099–1102 CrossRef CAS .
  66. C. C. Ruan, Z. Liu, X. Li, X. Liu, L. J. Wang, H. Y. Pan, Y. N. Zheng, G. Z. Sun, Y. S. Zhang and L. X. Zhang, Molecules, 2010, 15, 2319–2325 Search PubMed .
  67. Y. N. Meng, X. G. Li, Y. N. Meng, J. Y. Wang, Y. N. Zheng and X. W. Yang, Chem. J. Chin. Univ., 2007, 28, 1316–1318 .
  68. J. T. Chen, H. Z. Li, D. Wang, Y. J. Zhang and C. R. Yang, Helv. Chim. Acta, 2006, 89, 1442–1448 CrossRef CAS .
  69. M. Yoshikawa, T. Morikawa, Y. Kashima, K. Ninomiya and H. Matsuda, J. Nat. Prod., 2003, 66, 922–927 CrossRef CAS .
  70. K. Zou, S. Zhu, M. R. Meselhy, C. Tohda, S. Q. Cai and K. Komatsu, J. Nat. Prod., 2002, 65, 1288–1292 CrossRef CAS .
  71. M. Yoshikawa, S. Sugimoto, S. Nakamura and H. Matsuda, Chem. Pharm. Bull., 2007, 55, 571–576 CrossRef CAS .
  72. J. R. Wang, Y. Yamasaki, T. Tanaka, I. Kouno and Z. H. Jiang, Molecules, 2009, 14, 2087–2094 Search PubMed .
  73. F. Qiu, Z. Z. Ma, S. X. Xu, X. S. Yao, C. T. Che and Y. J. Chen, J. Asian Nat. Prod. Res., 2001, 3, 235–240 CrossRef CAS .
  74. H. T. Nguyen, G. Y. Song, V. M. Chau, V. K. Phan, L. G. Jin, H. J. Boo, H. K. Kang and Y. H. Kim, Chem. Pharm. Bull., 2010, 58, 1111–1115 CrossRef .
  75. N. Komakine, M. Okasaka, Y. Takaishi, K. Kawazoe, K. Murakami and Y. Yamada, J. Nat. Med., 2006, 60, 135–137 Search PubMed .
  76. N. H. Tung, G. Y. Song, Y. J. Park and Y. H. Kim, Chem. Pharm. Bull., 2009, 57, 1412–1414 CrossRef .
  77. J. H. Wang, W. Li, Y. Sha, Y. Tezuka, S. Kadota and X. Li, J. Asian Nat. Prod. Res., 2001, 3, 123–130 CrossRef CAS .
  78. J. Huang, X. H. Tang, T. Ikejima, X. J. Sun, X. B. Wang, R. G. Xi and L. J. Wu, Arch. Pharmacal Res., 2008, 31, 323–329 Search PubMed .
  79. J. Wang, Y. Sha, W. Li, Y. Tezuka, S. Kadota and X. Li, J. Asian Nat. Prod. Res., 2001, 3, 293–297 CrossRef CAS .
  80. Y. K. Qiu, D. Q. Dou, L. P. Cai, H. P. Jiang, T. G. Kang, B. Y. Yang, H. X. Kuang and M. Z. C. Li, Fitoterapia, 2009, 80, 219–222 CrossRef CAS .
  81. Y. Zhao, W. Wang, L. Han, E. R. Rayburn, D. L. Hill, H. Wang and R. Zhang, Med. Chem., 2007, 3, 51–60 CrossRef CAS .
  82. L. N. Tao, Q. Meng, J. Y. Yin, R. Xing and H. R. Guo, Chin. Chem. Lett., 2009, 20, 687–689 CrossRef CAS .
  83. X. W. Yang, L. Y. Li, J. M. Tian, Z. W. Zhang, J. M. Ye and W. F. Gu, Chin. Chem. Lett., 2000, 11, 909–912 CAS .
  84. I. H. Park, S. B. Han, J. M. Kim, L. Z. Piao, S. W. Kwon, N. Y. Kim, T. L. Kang, M. K. Park and J. H. Park, Arch. Pharmacal Res., 2002, 25, 837–841 Search PubMed .
  85. D. Dou, W. Li, N. Guo, R. Fu, Y. Pei, K. Koike and T. Nikaido, Chem. Pharm. Bull., 2006, 54, 751–753 CrossRef CAS .
  86. L. B. Wang, Z. H. Wu, H. Y. Gao, J. Huang, B. H. Sun and L. J. Wu, Chin. Chem. Lett., 2008, 19, 837–840 CrossRef CAS .
  87. L. J. Wu, L. B. Wang, H. Y. Gao, B. Wu, X. M. Song and Z. S. Tang, Fitoterapia, 2007, 78, 556–560 CrossRef CAS .
  88. H. Z. Li, R. W. Teng and C. R. Yang, Chin. Chem. Lett., 2001, 12, 59–62 CAS .
  89. H. J. Zhang, Z. Z. Lu, G. T. Tan, S. X. Qiu, N. R. Farnsworth, J. M. Pezzuto and H. H. S. Fong, Tetrahedron Lett., 2002, 43, 973–977 CrossRef CAS .
  90. Y. Wei, C. M. Ma and M. Hattori, Phytochem. Lett., 2009, 2, 63–66 Search PubMed .
  91. J. H. Liu, C. S. Lee, K. M. Leung, Z. K. Yan, B. H. Shen, Z. Z. Zhao and Z. H. Jiang, J. Agric. Food Chem., 2007, 55, 8830–8835 CrossRef CAS .
  92. M. C. Yang, H. C. Kwon, Y. J. Kim, K. R. Lee and H. O. Yang, J. Nat. Prod., 2010, 73, 801–805 CrossRef CAS .
  93. H. H. Chan, H. D. Sun, M. V. B. Reddy and T. S. Wu, Phytochemistry, 2010, 71, 1360–1364 CrossRef CAS .
  94. S. W. Lee, K. Kim, M. C. Rho, M. Y. Chung, Y. H. Kim, S. Lee, H. S. Lee and Y. K. Kim, Planta Med., 2004, 70, 197–200 CrossRef CAS .
  95. K. Hirakura, K. Sugama, M. Morita, K. Nakajima, H. Sasaki, M. Okada and H. Sato, Heterocycles, 2000, 53, 2451–2457 CrossRef CAS .
  96. K. Rittenbach, R. Ilarraza, Y. Wu, F. Davoine and D. J. Adamko, Pharm. Biol., 2009, 47, 17–17 .
  97. R. C. Y. Choi, J. T. T. Zhu, K. W. Leung, G. K. Y. Chu, H. Q. Xie, V. P. Chen, K. Y. Z. Zheng, D. T. W. Lau, T. T. X. Dong, P. C. Y. Chow, Y. F. Han, Z. T. Wang and K. W. K. Tsim, J. Alzheimers Dis., 2010, 19, 795–811 CAS .
  98. C. D. Liu, J. Chen and J. H. Wang, Chem. Nat. Compd., 2009, 45, 808–810 CrossRef CAS .
  99. M. V. Naval, M. P. Gomez-Serranillos, M. E. Carretero and C. De Arce, J. Chromatogr., A, 2006, 1121, 242–247 CrossRef CAS .
  100. J. Y. Wang, X. G. Li and X. W. Yang, J. Asian Nat. Prod. Res., 2006, 8, 605–608 CrossRef CAS .
  101. Y. Liu, J. L. Zhou, P. Liu, S. Sun and P. Li, J. Chromatogr. A, 1217, pp. 5239–5245 Search PubMed.
  102. D. Y. Hui, J. Nutr., 1998, 128, 2052–2057 CAS .
  103. J. T. Xie, H. H. Aung, J. A. Wu, A. S. Attele and C. S. Yuan, Am. J. Chin. Med., 2002, 30, 187–194 Search PubMed .
  104. A. B. Wang, C. Z. Wang, J. A. Wu, J. Osinski and C. S. Yuan, Phytochem. Anal., 2005, 16, 272–277 CrossRef CAS .
  105. X. H. Zhang, H. L. Liu and J. M. Chen, J. Chromatogr. Sci., 2005, 43, 47–51 CAS .
  106. S. P. Ip and C. T. Che, J. Chromatogr., A, 2006, 1135, 241–244 CrossRef CAS .
  107. N. Fukuda, H. Tanaka and Y. Shoyama, Biol. Pharm. Bull., 1999, 22, 219–220 CAS .
  108. N. Fukuda, H. Tanaka and Y. Shoyama, Cytotechnology, 2000, 34, 197–204 CrossRef CAS .
  109. O. Morinaga, N. Fukuda, H. Tanaka and Y. Shoyama, Glycobiology, 2005, 15, 1061–1066 CrossRef CAS .
  110. O. Morinaga, H. Tanaka and Y. Shoyama, J. Chromatogr., B, 2006, 830, 100–104 CrossRef CAS .
  111. S. J. Shan, H. Tanaka and Y. Shoyama, Anal. Chem., 2001, 73, 5784–5790 CrossRef CAS .
  112. N. Fukuda, H. Tanaka and Y. Shoyama, Analyst, 2000, 125, 1425–1429 RSC .
  113. H. Tanaka, N. Fukuda and Y. Shoyama, Phytochem. Anal., 2006, 17, 46–55 CrossRef CAS .
  114. H. Tanaka, N. Fukuda and Y. Shoyama, J. Agric. Food Chem., 2007, 55, 3783–3787 CrossRef CAS .
  115. H. Tanaka, N. Fukuda, S. Yahara, S. Isoda, C. S. Yuan and Y. Shoyama, Phytother. Res., 2005, 19, 255–258 CrossRef CAS .
  116. W. Putalun, N. Fukuda, H. Tanaka and Y. Shoyama, Anal. Bioanal. Chem., 2004, 378, 1338–1341 CrossRef CAS .
  117. C. Seger and S. Sturm, J. Proteome Res., 2007, 6, 480–497 CrossRef CAS .
  118. F. van der Kooy, F. Maltese, Y. H. Choi, H. K. Kim and R. Verpoorte, Planta Med., 2009, 75, 763–775 CrossRef CAS .
  119. B. Rasmussen, O. Cloarec, H. R. Tang, D. Staerk and J. W. Jaroszewski, Planta Med., 2006, 72, 556–563 CrossRef CAS .
  120. J. Kang, S. Lee, S. Kang, H. N. Kwon, J. H. Park, S. W. Kwon and S. Park, Arch. Pharmacal Res., 2008, 31, 330–336 Search PubMed .
  121. W. N. Lin, H. Y. Lu, M. S. Lee, S. Y. Yang, H. J. Chen, Y. S. Chang and W. T. Chang, Am. J. Chin. Med., 2010, 38, 205–218 Search PubMed .
  122. S. Y. Yang, H. K. Kim, A. W. M. Lefeber, C. Erkelens, N. Angelova, Y. H. Choi and R. Verpoorte, Planta Med., 2006, 72, 364–369 CrossRef CAS .
  123. K. Y. L. Yap, S. Y. Chan and C. S. Lim, J. Biomed. Sci., 2007, 14, 265–273 CrossRef CAS .
  124. K. Y. L. Yap, T. K. H. Lai, S. Y. Chan and C. S. Lim, J. AOAC Int., 2009, 92, 672–679 CAS .
  125. Y. J. Lu, Y. L. Qu, Z. Q. Cao and M. Song, Spectrosc. Spect. Anal., 2006, 26, 1457–1459 Search PubMed .
  126. X. J. Jin, Y. Zhang, Y. F. Xie, Q. Cong and B. Zhao, Spectrosc. Spect. Anal., 2009, 29, 656–660 Search PubMed .
  127. D. T. Li, C. G. Cheng, Z. X. Du, Y. Q. He and L. C. Kong, Spectrosc. Spect. Anal., 2006, 26, 2186–2189 Search PubMed .
  128. G. H. Lu, Q. Zhou, S. Q. Sun, K. S. Y. Leung, H. Zhang and Z. Z. Zhao, J. Mol. Struct., 2008, 883, 91–98 CrossRef .
  129. D. Liu, Y. G. Li, H. Xu, S. Q. Sun and Z. T. Wang, J. Mol. Struct., 2008, 883, 228–235 CrossRef .
  130. Y. M. Li, S. Q. Sun, Q. Zhou, Z. Qin, J. X. Tao, J. Wang and X. Fang, Vib. Spectrosc., 2004, 36, 227–232 CrossRef CAS .
  131. Y. L. Zhang, J. B. Chen, Y. Lei, Q. Zhou, S. Q. Sun and I. Noda, J. Mol. Struct., 2010, 974, 94–102 CrossRef CAS .
  132. J. J. Mao and J. W. Xu, Spectrochim. Acta, Part A, 2006, 65, 497–500 CrossRef .
  133. M. Baranska, H. Schulz and L. P. Christensen, J. Agric. Food Chem., 2006, 54, 3629–3635 CrossRef CAS .
  134. A. Ludwiczuk, T. Wolski and S. Berbec, J. Planar Chromatogr.–Mod. TLC, 2002, 15, 147–150 CrossRef CAS .
  135. T. Ohno, E. Mikami and H. Oka, J. Nat. Med., 2006, 60, 141–145 Search PubMed .
  136. Y. Q. Qiu, X. Lu, T. Pang, C. F. Ma, X. Li and G. W. Xu, J. Sep. Sci., 2008, 31, 3451–3457 CrossRef CAS .
  137. X. Di, R. A. Shellie, P. J. Marriott and C. W. Huie, J. Sep. Sci., 2004, 27, 451–458 CrossRef CAS .
  138. X. Y. Wang, M. M. Su, Y. P. Qiu, Y. Ni, T. Zhao, M. M. Zhou, A. H. Zhao, S. L. Yang, L. P. Zhao and W. Jia, J. Proteome Res., 2007, 6, 3449–3455 CrossRef CAS .
  139. W. Lim, K. W. Mudge and F. Vermeylen, J. Agric. Food Chem., 2005, 53, 8498–8505 CrossRef CAS .
  140. W. K. Li and J. F. Fitzloff, J. Liq. Chromatogr. Relat. Technol., 2002, 25, 17–27 CrossRef CAS .
  141. T. W. D. Chan, P. P. H. But, S. W. Cheng, I. M. Y. Kwok, F. W. Lau and H. X. Xu, Anal. Chem., 2000, 72, 1281–1287 CrossRef CAS .
  142. H. L. Koh, A. J. Lau and E. C. Y. Chan, Rapid Commun. Mass Spectrom., 2005, 19, 1237–1244 CrossRef CAS .
  143. D. W. Lou, Y. Saito, P. K. Zarzycki, M. Ogawa and K. Jinno, Anal. Bioanal. Chem., 2006, 385, 96–104 CrossRef CAS .
  144. N. S. Quiming, N. L. Denola, Y. Saito and K. Jinno, J. Sep. Sci., 2008, 31, 1550–1563 CrossRef CAS .
  145. N. S. Quiming, N. L. Denola, A. B. Soliev, Y. Saito and K. Jinno, Anal. Bioanal. Chem., 2007, 389, 1477–1488 CrossRef CAS .
  146. M. Dan, M. M. Su, X. F. Gao, T. Zhao, A. H. Zhao, G. X. Xie, Y. P. Qiu, M. M. Zhou, Z. Liu and W. Jia, Phytochemistry, 2008, 69, 2237–2244 CrossRef CAS .
  147. G. X. Xie, Y. Ni, M. M. Su, Y. Y. Zhang, A. H. Zhao, X. F. Gao, Z. Liu, P. G. Xiao and W. Jia, Metabolomics, 2008, 4, 248–260 CrossRef CAS .
  148. G. X. Xie, R. Plumb, M. M. Su, Z. H. Xu, A. H. Zhao, M. F. Qiu, X. B. Long, Z. Liu and W. Jia, J. Sep. Sci., 2008, 31, 1015–1026 CrossRef CAS .
  149. I. Glöckl, M. Veit and G. Blaschke, Planta Med., 2002, 68, 158–161 CrossRef CAS .
  150. J. Cao, L. W. Qi, J. Chen and P. Li, Electrophoresis, 2008, 29, 4422–4430 CrossRef CAS .
  151. J. Cao, L. Yi, P. Li and Y. X. Chang, J. Chromatogr., A, 2009, 1216, 5608–5613 CrossRef CAS .
  152. J. Cao, B. Li, Y. X. Chang and P. Li, Electrophoresis, 2009, 30, 1372–1379 CrossRef CAS .
  153. R. A. Shellie, P. J. Marriott and C. W. Huie, J. Sep. Sci., 2003, 26, 1185–1192 CrossRef CAS .
  154. E. Reich, A. Schibli and A. DeBatt, J. AOAC Int., 2008, 91, 13–20 CAS .
  155. P. Xie, S. Chen, Y. Z. Liang, X. Wang, R. Tian and R. Upton, J. Chromatogr., A, 2006, 1112, 171–180 CrossRef CAS .
  156. R. J. Vanhaelen-Fastre, M. L. Faes and M. H. Vanhaelen, J. Chromatogr., A, 2000, 868, 269–276 CrossRef CAS .
  157. C. Kevers, P. Jacques, T. Gaspar, P. Thonart and J. Dommes, J. Chromatogr. Sci., 2004, 42, 554–558 CAS .
  158. A. M. A. El-Aty, I. K. Kim, M. R. Kim, C. Lee and J. H. Shim, Biomed. Chromatogr., 2008, 22, 556–562 CrossRef .
  159. J. W. Wong, K. Zhang, K. Tech, D. G. Hayward, A. J. Krynitsky, I. Cassias, F. J. Schenck, K. Banerjee, S. Dasgupta and D. Brown, J. Agric. Food Chem., 2010, 58, 5884–5896 CrossRef CAS .
  160. D. G. Hayward and J. W. Wong, Anal. Chem., 2009, 81, 5716–5723 CrossRef CAS .
  161. X. Y. Sun, Y. P. Zheng, D. H. Lin, H. Zhang, F. Zhao and C. S. Yuan, Am. J. Chin. Med., 2009, 37, 589–596 Search PubMed .
  162. N. S. Quiming, N. L. Denola, A. B. Soliev, Y. Saito and K. Jinno, Chromatographia, 2007, 66, 5–11 CrossRef CAS .
  163. S. Louw, A. S. Pereira, F. Lynen, M. Hanna-Brown and P. Sandra, J. Chromatogr., A, 2008, 1208, 90–94 CrossRef CAS .
  164. L. Li, G. A. Luo, Q. L. Liang, P. Hu and Y. M. Wang, J. Pharm. Biomed. Anal., 2010, 52, 66–72 CrossRef .
  165. D. Muth, E. Marsden-Edwards, P. Kachlicki and M. Stobiecki, Phytochem. Anal., 2008, 19, 444–452 CrossRef CAS .
  166. D. Guillarme, J. Schappler, S. Rudaz and J. L. Veuthey, TrAC, Trends Anal. Chem., 2010, 29, 15–27 CrossRef CAS .
  167. J. Guan, C. M. Lai and S. P. Li, J. Pharm. Biomed. Anal., 2007, 44, 996–1000 CrossRef CAS .
  168. G. F. Deng, D. L. Wang, M. X. Meng, F. Hu and T. W. Yao, J. Chromatogr., B, 2009, 877, 2113–2122 CrossRef CAS .
  169. X. Y. Wang, T. Zhao, X. F. Gao, M. Dan, M. M. Zhou and W. Jia, Anal. Chim. Acta, 2007, 594, 265–273 CrossRef CAS .
  170. L. W. Qi, C. Y. Chen and P. Li, Rapid Commun. Mass Spectrom., 2009, 23, 3227–3242 CrossRef CAS .
  171. E. H. Liu, L. W. Qi, B. Li, Y. B. Peng, P. Li, C. Y. Li and J. Cao, Rapid Commun. Mass Spectrom., 2009, 23, 119–130 CrossRef CAS .
  172. T. J. Pappas, M. Gayton-Ely and L. A. Holland, Electrophoresis, 2005, 26, 719–734 CrossRef CAS .
  173. C. W. Huie, Electrophoresis, 2006, 27, 60–75 CrossRef CAS .
  174. J. H. Qin, F. C. Leung, Y. S. Fung, D. R. Zhu and B. C. Lin, Anal. Bioanal. Chem., 2005, 381, 812–819 CrossRef CAS .
  175. S. F. Wang, S. Ye and Y. Y. Cheng, J. Chromatogr., A, 2006, 1109, 279–284 CrossRef CAS .
  176. L. Ciesla and M. Waksmundzka-Hajnos, J. Chromatogr., A, 2009, 1216, 1035–1052 CrossRef CAS .
  177. R. K. Gilpin and C. S. Gilpin, Anal. Chem., 2009, 81, 4679–4694 CrossRef CAS .
  178. L. W. Qi, Q. T. Yu, P. Li, S. L. Li, Y. X. Wang, L. H. Sheng and L. Yi, J. Chromatogr., A, 2006, 1134, 162–169 CrossRef CAS .
  179. J. Cao, Y. J. Wei, L. W. Qi, P. Li, Z. M. Qian, H. W. Luo, J. Chen and J. Zhao, Biomed. Chromatogr., 2008, 22, 164–172 CrossRef CAS .
  180. S. N. Kim, Y. W. Ha, H. Shin, S. H. Son, S. J. Wu and Y. S. Kim, J. Pharm. Biomed. Anal., 2007, 45, 164–170 CrossRef CAS .
  181. W. K. Li and J. F. Fitzloff, J. Liq. Chromatogr. Relat. Technol., 2002, 25, 29–41 CrossRef CAS .
  182. T. Vehovec and A. Obreza, J. Chromatogr., A, 2010, 1217, 1549–1556 CrossRef CAS .
  183. C. C. Bai, S. Y. Han, X. Y. Chai, Y. Jiang, P. Li and P. F. Tu, J. Liq. Chromatogr. Relat. Technol., 2009, 32, 242–260 CrossRef CAS .
  184. L. Wang, W. S. He, H. X. Yan, Y. Jiang, K. S. Bi and P. F. Tu, Chromatographia, 2009, 70, 603–608 CrossRef CAS .
  185. K. S. Y. Leung, K. Chan, A. Bensoussan and M. J. Munroe, Phytochem. Anal., 2007, 18, 146–150 CrossRef CAS .
  186. X. Ma, H. B. Xiao and X. M. Liang, Chromatographia, 2006, 64, 31–36 CrossRef .
  187. X. Q. Ma, X. M. Liang, Q. Xu, X. Z. Zhang and H. B. Xiao, Phytochem. Anal., 2005, 16, 181–187 CrossRef CAS .
  188. Y. Liu, J. Li, J. He, Z. Abliz, J. Qu, S. Yu, S. Ma, J. Liu and D. Du, Rapid Commun. Mass Spectrom., 2009, 23, 667–679 CrossRef CAS .
  189. S. Liu, M. Cui, Z. Liu, F. Song and W. Mo, J. Am. Soc. Mass Spectrom., 2004, 15, 133–141 CrossRef CAS .
  190. B. D. Sloley, Y. C. J. Lin, D. Ridgway, H. A. Semple, Y. K. Tam, R. T. Coutts, R. Lobenberg and N. Tam-Zaman, J. AOAC Int., 2006, 89, 16–21 CAS .
  191. C. H. Xia, G. J. Wang, R. G. Sun, H. P. Hao, Y. Q. Xiong, S. H. Gu, L. L. Shang and C. N. Zheng, J. Chromatogr., B, 2008, 862, 72–78 CrossRef CAS .
  192. J. L. Zhou, P. Li, H. J. Li, Y. Jiang, M. T. Ren and Y. Liu, J. Chromatogr., A, 2008, 1177, 126–137 CrossRef CAS .
  193. J. W. Wong, M. K. Hennessy, D. G. Hayward, A. J. Krynitsky, I. Cassias and F. J. Schenck, J. Agric. Food Chem., 2007, 55, 1117–1128 CrossRef CAS .
  194. G. C. Kite, M. J. R. Howes, C. J. Leon and M. S. J. Simmonds, Rapid Commun. Mass Spectrom., 2003, 17, 238–244 CrossRef CAS .
  195. W. C. S. Cho, T. T. Yip, W. S. Chung, S. K. W. Lee, A. W. N. Leung, C. H. K. Cheng and K. K. M. Yue, J. Ethnopharmacol., 2006, 108, 272–279 CrossRef CAS .
  196. S. L. Li, S. F. Lai, J. Z. Song, C. F. Qiao, X. Liu, Y. Zhou, H. Cai, B. C. Cai and H. X. Xu, J. Pharm. Biomed. Anal., 2010, 53, 946–957 CrossRef CAS .
  197. L. Xia, H. L. Liu, P. Li, J. L. Zhou, L. W. Qi, L. Yi and J. Chen, J. Sep. Sci., 2008, 31, 3156–3169 CrossRef CAS .
  198. H. Kong, M. Wang, K. Venema, A. Maathuis, R. van der Heijden, J. van der Greef, G. Xu and T. Hankemeier, J. Chromatogr., A, 2009, 1216, 2195–2203 CrossRef CAS .
  199. K. Tanaka, M. Kubota, S. Zhu, U. Sankawa and K. Komatsu, Nat. Prod. Commun., 2007, 2, 625–632 CAS .
  200. Y. W. Ha, K. S. Ahn, J. C. Lee, S. H. Kim, B. C. Chung and M. H. Choi, Anal. BioAnal. Chem., 396, pp. 3017–3025 Search PubMed.
  201. Q. C. Ji, M. R. Harkey, G. L. Henderson, M. E. Gershwin, J. S. Stern and R. M. Hackman, Phytochem. Anal., 2001, 12, 320–326 CrossRef CAS .
  202. H. Hao, N. Cui, G. Wang, B. Xiang, Y. Liang, X. Xu, H. Zhang, J. Yang, C. Zheng, L. Wu, P. Gong and W. Wang, Anal. Chem., 2008, 80, 8187–8194 CrossRef CAS .
  203. C. Zheng, H. Hao, X. Wang, X. Wu, G. Wang, G. Sang, Y. Liang, L. Xie, C. Xia and X. Yao, J. Mass Spectrom., 2009, 44, 230–244 CrossRef CAS .
  204. Q. Wu, Q. Yuan, E. H. Liu, L. W. Qi, Z. M. Bi and P. Li, Biomed. Chromatogr., 2009 Search PubMed .
  205. R. Hu, J. Zhao, L. W. Qi, P. Li, S. L. Jing and H. J. Li, Rapid Commun. Mass Spectrom., 2009, 23, 1619–1635 CrossRef CAS .
  206. L. W. Qi, X. D. Wen, J. Cao, C. Y. Li, P. Li, L. Yi, Y. X. Wang, X. L. Cheng and X. X. Ge, Rapid Commun. Mass Spectrom., 2008, 22, 2493–2509 CrossRef CAS .
  207. L. W. Qi, J. Cao, P. Li, Q. T. Yu, X. D. Wen, Y. X. Wang, C. Y. Li, K. D. Bao, X. X. Ge and X. L. Cheng, J. Chromatogr., A, 2008, 1203, 27–35 CrossRef CAS .
  208. B. Sritularak, O. Morinaga, C. S. Yuan, Y. Shoyama and H. Tanaka, J. Nat. Med., 2009, 63, 360–363 Search PubMed .
  209. O. Morinaga, T. Uto, C. S. Yuan, H. Tanaka and Y. Shoyama, Fitoterapia, 2010, 81, 284–288 CrossRef CAS .
  210. C. Z. Wang, M. Ni, S. Sun, X. L. Li, H. He, S. R. Mehendale and C. S. Yuan, J. Agric. Food Chem., 2009, 57, 2363–2367 CrossRef CAS .
  211. P. S. Xie and A. Y. Leung, J. Chromatogr., A, 2009, 1216, 1933–1940 CrossRef CAS .
  212. E. J. Lee, R. Shaykhutdinov, A. M. Weljie, H. J. Vogel, P. J. Facchini, S. U. Park, Y. K. Kim and T. J. Yang, J. Agric. Food Chem., 2009, 57, 7513–7522 CrossRef CAS .
  213. W. K. Li, C. G. Gu, H. J. Zhang, D. V. C. Awang, J. F. Fitzloff, H. H. S. Fong and R. B. van Breemen, Anal. Chem., 2000, 72, 5417–5422 CrossRef CAS .
  214. E. M. Schlag and M. S. McIntosh, Phytochemistry, 2006, 67, 1510–1519 CrossRef CAS .
  215. C. C. Hon, Y. C. Chow, F. Y. Zeng and F. C. C. Leung, Acta Pharmacol. Sin., 2003, 24, 841–846 Search PubMed .
  216. K. S. Cheung, H. S. Kwan, P. P. H. But and P. C. Shaw, J. Ethnopharmacol., 1994, 42, 67–69 CrossRef CAS .
  217. J. Y. Um, H. S. Chung, M. S. Kim, H. J. Na, H. J. Kwon, J. J. Kim, K. M. Lee, S. J. Lee, J. P. Lim, K. R. Do, W. J. Hwang, Y. S. Lyu, N. H. An and H. M. Kim, Biol. Pharm. Bull., 2001, 24, 872–875 CrossRef CAS .
  218. J. Wang, W. Y. Ha, F. N. Ngan, P. P. H. But and P. C. Shaw, Planta Med., 2001, 67, 781–783 CrossRef CAS .
  219. I. S. H. Ho and F. C. Leung, Mol. Genet. Genomics, 2002, 266, 951–961 CrossRef CAS .
  220. W. Y. Ha, P. C. Shaw, J. Liu, F. C. F. Yau and J. Wang, J. Agric. Food Chem., 2002, 50, 1871–1875 CrossRef CAS .
  221. H. Fushimi, K. Komatsu, M. Isobe and T. Namba, Biol. Pharm. Bull., 1997, 20, 765–769 CAS .
  222. W. Y. Ha, F. C. F. Yau, P. P. H. But, J. Wang and P. C. Shaw, Planta Med., 2001, 67, 587–589 CrossRef CAS .
  223. S. Zhu, H. Fushimi, S. Q. Cai and K. Komatsu, Planta Med., 2003, 69, 647–653 CrossRef CAS .
  224. S. Zhu, H. Fushimi, S. Cai and K. Komatsu, Planta Med., 2004, 70, 189–192 CrossRef CAS .
  225. Y. Diao, X. M. Lin, C. L. Liao, C. Z. Tang, Z. J. Chen and Z. L. Hu, Planta Med., 2009, 75, 557–560 CrossRef CAS .
  226. Y. Sasaki, K. Komatsu and S. Nagumo, Biol. Pharm. Bull., 2008, 31, 1806–1808 CrossRef CAS .
  227. K. T. Lu, H. C. Lee, F. S. Liu, C. F. Lo and J. H. Lin, J. Food Drug Anal., 2010, 18, 58–63 Search PubMed .
  228. X. M. Cui, C. K. Lo, K. L. Yip, T. T. X. Dong and K. W. K. Tsim, Planta Med., 2003, 69, 584–586 CrossRef CAS .
  229. A. Mathur, A. K. Mathur, R. S. Sangwan, A. Gangwar and G. C. Uniyal, Genet. Resour. Crop Evol., 2003, 50, 245–252 Search PubMed .
  230. D. P. Bai, J. Brandle and R. Reeleder, Genome, 1997, 40, 111–115 CrossRef CAS .
  231. W. Lim and K. Mudgez, Hortscience, 2004, 39, 889–889 Search PubMed .
  232. W. Lim, K. W. Mudge and L. A. Weston, Planta Med., 2007, 73, 71–76 CrossRef CAS .
  233. C. Z. Wang, P. Li, J. Y. Ding, G. Q. Jin and C. S. Yuan, Planta Med., 2005, 71, 384–386 CrossRef CAS .
  234. Y. E. Choi, C. H. Ahn, B. B. Kim and E. S. Yoon, Biol. Pharm. Bull., 2008, 31, 135–138 CrossRef CAS .
  235. G. D. Reunova, I. L. Kats, T. I. Muzarok and Y. N. Zhuravlev, Russ. J. Genet., 2010, 46, 938–947 Search PubMed .
  236. N. V. Dan, N. Ramchiary, S. R. Choi, T. S. Uhm, T. J. Yang, I. O. Ahn and Y. P. Lim, Conserv. Genet., 2010, 11, 1223–1225 Search PubMed .
  237. K. H. Ma, A. Dixit, Y. C. Kim, D. Y. Lee, T. S. Kim, E. G. Cho and Y. J. Park, Conserv. Genet., 2007, 8, 1507–1509 Search PubMed .
  238. Y. S. Shin, K. H. Bang, D. S. In, O. T. Kim, D. Y. Hyun, I. O. Ahn, B. C. Ku, S. W. Kim, N. S. Seong, S. W. Cha, D. Lee and H. K. Choi, Arch. Pharmacal Res., 2007, 30, 1625–1628 Search PubMed .
  239. Z. M. Qian, J. Lu, Q. P. Gao and S. P. Li, J. Chromatogr., A, 2009, 1216, 3825–3830 CrossRef CAS .
  240. V. A. Assinewe, B. R. Baum, D. Gagnon and J. T. Arnason, J. Agric. Food Chem., 2003, 51, 4549–4553 CrossRef CAS .
  241. Y. Wang, J. Y. Pan, X. Y. Xiao, R. C. Lin and Y. Y. Cheng, Phytochem. Anal., 2006, 17, 424–430 CrossRef CAS .
  242. S. Perez and D. Barcelo, TrAC, Trends Anal. Chem., 2007, 26, 494–514 CrossRef CAS .
  243. L. W. Qi, X. J. Gu, P. Li, Y. Liang, H. Hao and G. Wang, Rapid Commun. Mass Spectrom., 2009, 23, 2151–2160 CrossRef CAS .
  244. N. Fuzzati, B. Gabetta, K. Jayakar, R. Pace and F. Peterlongo, J. Chromatogr., A, 1999, 854, 69–79 CrossRef CAS .
  245. I. Kitagawa, T. Taniyama, T. Hayashi and M. Yoshikawa, Chem. Pharm. Bull., 1983, 31, 3353–3356 CAS .
  246. H. Yamaguchi, R. Kasai, H. Matsuura, O. Tanaka and T. Fuwa, Chem. Pharm. Bull., 1988, 36, 3468–3473 CAS .
  247. G. X. Ren and F. Chen, J. Agric. Food Chem., 1999, 47, 1501–1505 CrossRef CAS .
  248. Y. H. Chang and P. K. W. Ng, J. Agric. Food Chem., 2009, 57, 2356–2362 CrossRef CAS .
  249. W. C. Chuang, H. K. Wu, S. J. Sheu, S. H. Chiou, H. C. Chang and Y. P. Chen, Planta Med., 1995, 61, 459–465 CAS .
  250. W. A. Court, J. G. Hendel and J. Elmi, J. Chromatogr., A, 1996, 755, 11–17 CrossRef CAS .
  251. L. P. Christensen, M. Jensen and U. Kidmose, J. Agric. Food Chem., 2006, 54, 8995–9003 CrossRef CAS .
  252. V. J. Davidson, X. Li and R. B. Brown, J. Food Eng., 2004, 63, 369–373 CrossRef .
  253. A. J. Lau, S. O. Woo and H. L. Koh, J. Chromatogr., A, 2003, 1011, 77–87 CrossRef CAS .
  254. A. J. Lau, D. F. Toh, T. K. Chua, Y. K. Pang, S. O. Woo and H. L. Koh, J. Ethnopharmacol., 2009, 125, 380–386 CrossRef CAS .
  255. Y. J. Lee, H. Y. Kim, K. S. Kang, J. G. Lee, T. Yokozawa and J. H. Park, Bioorg. Med. Chem. Lett., 2008, 18, 4515–4520 CrossRef CAS .
  256. S. Su, L. W. Qi, G. J. Du, S. R. Mehendale, C. Z. Wang and C. S. Yuan, Food Chem., 2011, 125, 1299–1305 CrossRef .
  257. S. Chan, M. F. Kong, Y. C. Wong, S. K. Wong and D. W. Sin, J. Agric. Food Chem., 2007, 55, 3339–3345 CrossRef CAS .
  258. S. H. Sohn, S. K. Kim, H. G. Kang and J. J. Wee, J. Chromatogr., A, 2004, 1042, 163–168 CrossRef CAS .
  259. F. E. Ahmed, TrAC, Trends Anal. Chem., 2001, 20, 649–661 CrossRef CAS .
  260. L. W. Chu, J. C. Yan, D. Chen, Z. G. Hou and Z. X. Fan, Chin. J. Anal. Chem., 2006, 34, 1482–1486 CAS .
  261. Y. S. Park, A. M. Abd El-Aty, J. H. Choi, S. K. Cho, D. H. Shin and J. H. Shim, Biomed. Chromatogr., 2007, 21, 29–39 CrossRef CAS .
  262. H. W. Wen, M. F. Hsieh, Y. T. Wang, H. P. Chung, P. C. Hsieh, I. H. Lin and F. I. Chou, J. Hazard. Mater., 2010, 176, 280–287 CrossRef CAS .
  263. L. Quan, S. F. Li, S. J. Tian, H. Xu, A. Q. Lin and L. Gu, Chromatographia, 2004, 59, 89–93 .
  264. C. Quan, Y. G. Shang, S. F. Li, S. K. Tang, T. Huang and X. Fang, J. Taiwan Inst. Chem. Eng., 2010, 41, 44–48 Search PubMed .
  265. C. Quan, S. F. Li, S. J. Tian, H. Xu, A. Q. Lin and L. Gu, J. Supercrit. Fluids, 2004, 31, 149–157 CrossRef CAS .
  266. F. S. Dong, X. G. Liu, Y. Q. Zheng, J. Li, W. G. Zhang, X. Z. Zhang and C. J. Li, Chin. J Anal. Chem., 2008, 36, 1716–1720 CAS .
  267. L. J. Han, W. Li, L. K. Han, J. N. Hu, Z. Wang, W. C. Liu and Y. N. Zheng, Chem. Anal. (Warsaw), 2009, 54, 1433–1443 CAS .
  268. J. Li, F. S. Dong, X. G. Liu, Y. Q. Zheng, J. R. Yao and C. P. Zhang, Chromatographia, 2009, 69, 1113–1117 CAS .
  269. J. M. Durgnat, J. Heuser, D. Andrey and C. Perrin, Food Addit. Contam., Part A, 2005, 22, 1224–1230 Search PubMed .
  270. K. M. Joo, J. H. Lee, H. Y. Jeon, C. W. Park, D. K. Hong, H. J. Jeong, S. J. Lee, S. Y. Lee and K. M. Lim, J. Pharm. Biomed. Anal., 2010, 51, 278–283 CrossRef CAS .
  271. W. Wang, G. J. Wang, H. T. Xie, J. G. Sun, S. Zhao, X. L. Jiang, H. Li, H. Lv, M. J. Xu and R. Wang, J. Chromatogr., B, 2007, 852, 8–14 CrossRef CAS .
  272. X. Zeng, Y. H. Deng, Y. Feng, Y. M. Liu, L. Yang, Y. Huang, J. Sun, W. X. Liang and Y. Y. Guan, J. Clin. Pharmacol., 2010, 50, 285–292 CrossRef CAS .
  273. L. Yang, Y. H. Deng, S. J. Xu and X. Zeng, J. Chromatogr., B, 2007, 854, 77–84 CrossRef CAS .
  274. J. Sun, G. Wang, X. Haitang, L. Hao, P. Guoyu and I. Tucker, J. Pharm. Biomed. Anal., 2005, 38, 126–132 CAS .
  275. L. Lai, H. P. Hao, Y. T. Liu, C. N. Zheng, Q. Wang, G. J. Wang and X. J. Chen, Planta Med., 2009, 75, 797–802 CrossRef CAS .
  276. F. J. Gui, X. W. Yang, L. Y. Li and J. M. Tian, J. Chromatogr., B, 2007, 850, 1–6 CrossRef CAS .
  277. M. J. Xu, G. J. Wang, H. T. Xie, R. Wang, W. Wang, X. Y. Li and J. G. Sun, Anal. Lett., 2006, 39, 113–126 CrossRef CAS .
  278. H. T. Xie, G. J. Wang, J. G. Sun, I. Tucker, X. C. Zhao, Y. Y. Xie, H. Li, X. L. Jiang, R. Wang, M. J. Xu and W. Wang, J. Chromatogr., B, 2005, 818, 167–173 CrossRef CAS .
  279. Z. W. Cai, T. X. Qian, R. N. S. Wong and Z. H. Jiang, Anal. Chim. Acta, 2003, 492, 283–293 CrossRef CAS .
  280. Q. A. Zhao, X. Zheng, J. Jiang, H. Zhou and P. Hu, J. Chromatogr., B, 2010, 878, 2266–2273 CrossRef CAS .
  281. T. X. Qian, Z. W. Cai, R. N. S. Wong, N. K. Mak and Z. H. Jiang, J. Chromatogr., B, 2005, 816, 223–232 CrossRef CAS .
  282. T. X. Oian, Z. W. Cai, R. N. S. Wong and Z. H. Jiang, Rapid Commun. Mass Spectrom., 2005, 19, 3549–3554 CrossRef CAS .
  283. P. S. Lee, T. W. Song, J. H. Sung, D. C. Moon, S. Song and Y. B. Chung, Planta Med., 2006, 72, 204–210 CrossRef CAS .
  284. I. P. Paek, Y. Moon, J. Kim, H. Y. Ji, S. A. Kim, D. H. Sohn, J. B. Kim and H. S. Lee, Biopharm. Drug Dispos., 2006, 27, 39–45 CrossRef CAS .
  285. H. C. Ren, J. G. Sun, G. J. Wang, J. Y. A., H. T. Xie, W. B. Zha, B. Yan, F. Z. Sun, H. P. Hao, S. H. Gu, L. S. Sheng, F. Shao, J. Shi and F. Zhou, J. Pharm. Biomed. Anal., 2008, 48, 1476–1480 CrossRef CAS .
  286. D. Zhang, Y. W. Wang, J. B. Han, W. S. Yu, L. L. Deng, J. P. Fawcett, Z. Y. Liu and J. K. Gu, J. Chromatogr., B, 2009, 877, 581–585 CrossRef CAS .
  287. X. Zhang, D. Zhang, J. Xu, J. Gu and Y. Zhao, J. Chromatogr., B, 2007, 858, 65–70 CrossRef CAS .
  288. Q. F. Xu, X. L. Fang and D. F. Chen, J. Ethnopharmacol., 2003, 84, 187–192 CrossRef CAS .
  289. X. Y. Li, G. J. Wang, H. G. Sun, H. P. Hao, Y. Q. Xiong, B. Yan, Y. T. Zheng and L. S. Sheng, Biol. Pharm. Bull., 2007, 30, 847–851 CrossRef CAS .
  290. X. Wang, T. Zhao, X. Gao, M. Dan, M. Zhou and W. Jia, Anal. Chim. Acta, 2007, 594, 265–273 CrossRef CAS .
  291. L. Yang, S. J. Xu, Z. F. Wu, Y. M. Liu and X. Zeng, Phytother. Res., 2009, 23, 65–71 CrossRef CAS .
  292. Y. Liang, A. Kang, T. Xie, X. Zheng, C. Dai, H. Hao, J. A, L. Sheng, L. Xie and G. J. Wang, J. Chromatogr. A, 1217, pp. 4501–4506 Search PubMed.
  293. H. F. Liu, J. L. Yang, F. F. Du, X. M. Gao, X. T. Ma, Y. H. Huang, F. Xu, W. Niu, F. Q. Wang, Y. Mao, Y. Sun, T. Lu, C. X. Liu, B. L. Zhang and C. Li, Drug Metab. Dispos., 2009, 37, 2290–2298 CrossRef CAS .
  294. X. T. Duan, X. Y. Chen, M. H. Li, B. Song and D. F. Zhong, Drug Metab. Rev., 2006, 38, 109–109 .
  295. H. Hasegawa, J. Pharmacol. Sci., 2004, 95, 153–157 CAS .
  296. J. F. Cui, I. Bjorkhem and P. Eneroth, J. Chromatogr., B, 1997, 689, 349–355 CrossRef CAS .
  297. M. A. Tawab, U. Bahr, M. Karas, M. Wurglics and M. Schubert-Zsilavecz, Drug Metab. Dispos., 2003, 31, 1065–1071 CrossRef .
  298. Y. Gu, G. J. Wang, J. G. Sun, Y. W. Jia, W. Wang, M. J. Xu, T. Lv, Y. T. Zheng and Y. Sai, Food Chem. Toxicol., 2009, 47, 2257–2268 CrossRef CAS .
  299. P. S. Lee, J. Y. Han, T. W. Song, J. H. Sung, O. S. Kwon, S. Song and Y. B. Chung, Int. J. Pharm., 2006, 316, 29–36 CrossRef CAS .
  300. L. Li, Y. X. Sheng, J. L. Zhang, S. S. Wang and D. A. Guo, Biomed. Chromatogr., 2006, 20, 327–335 CrossRef CAS .
  301. J. Q. Ruan, W. I. Leong, R. Yan and Y. T. Wang, J. Agric. Food Chem., 2010, 58, 5770–5776 CrossRef CAS .
  302. G. T. Chen, M. Yang, Y. Song, Z. Q. Lu, J. Q. Zhang, H. L. Huang, S. H. Guan, L. J. Wu and D. A. Guo, Biomed. Chromatogr., 2008, 22, 779–785 CrossRef CAS .
  303. L. Yang, S. J. Xu, C. J. Liu and Z. J. Su, Anal. Bioanal. Chem., 2009, 395, 1441–1451 CrossRef CAS .
  304. H. Hasegawa, R. Suzuki, T. Nagaoka, Y. Tezuka, S. Kadota and I. Saiki, Biol. Pharm. Bull., 2002, 25, 861–866 CrossRef CAS .
  305. S. Rochfort, J. Nat. Prod., 2005, 68, 1813–1820 CrossRef CAS .
  306. M. J. Chen, M. M. Su, L. P. Zhao, J. Jiang, P. Liu, J. Y. Cheng, Y. J. Lai, Y. M. Liu and W. Jia, J. Proteome Res., 2006, 5, 995–1002 CrossRef CAS .
  307. M. J. Chen, L. P. Zhao and W. Jia, J. Proteome Res., 2005, 4, 2391–2396 CrossRef CAS .
  308. G. X. Xie, X. J. Zheng, X. Qi, Y. Cao, Y. Chi, M. M. Su, Y. Ni, Y. P. Qiu, Y. M. Liu, H. K. Li, A. H. Zhao and W. Jia, J. Proteome Res., 2010, 9, 125–133 CrossRef CAS .
  309. Y. Wang, J. Wang, M. Yao, X. Zhao, J. Fritsche, P. Schmitt-Kopplin, Z. Cai, D. Wan, X. Lu, S. Yang, J. Gu, H. U. Haring, E. D. Schleicher, R. Lehmann and G. Xu, Anal. Chem., 2008, 80, 4680–4688 CrossRef CAS .
  310. R. J. Tian, S. Y. Xu, X. Y. Lei, W. H. Jin, M. L. Ye and H. F. Zou, TrAC, Trends Anal. Chem., 2005, 24, 810–825 CrossRef CAS .
  311. Y. Duan, J. Zheng, V. Law and R. Nicholson, Eur. J. Pharmacol., 2006, 530, 9–14 CrossRef CAS .
  312. R. Nicholson, Y. Duan, J. Zheng and V. Law, Comp. Biochem. Physiol., Part A: Mol. Integr. Physiol., 2006, 143, S117–S117 CrossRef .
  313. J. L. Zhou, J. J. An, P. Li, H. J. Li, Y. Jiang and J. F. Cheng, J. Chromatogr., A, 2009, 1216, 2394–2403 CrossRef CAS .
  314. X. Y. Su, L. Kong, X. Y. Lei, L. H. Hu, M. L. Ye and H. F. Zou, Mini-Rev. Med. Chem., 2007, 7, 87–98 CrossRef CAS .
  315. X. D. Huang, L. A. Kong, X. Li, X. G. Chen, M. Guo and H. F. Zou, J. Chromatogr., B, 2004, 812, 71–84 CrossRef CAS .
  316. J. Wang, Z. G. Huang, H. Cao, Y. T. Wang, P. Hui, C. Hoo and S. P. Li, J. Sep. Sci., 2008, 31, 1173–1180 CrossRef CAS .
  317. Y. Liu, M. X. Xie, J. Kang and D. Zheng, Spectrochim. Acta, Part A, 2003, 59, 2747–2758 CrossRef .
  318. A. Dasgupta and M. A. Reyes, Am. J. Clin. Pathol., 2005, 124, 229–236 CrossRef CAS .

This journal is © The Royal Society of Chemistry 2011