You are on page 1of 29

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/225907237

Analysis of Catharanthus roseus alkaloids by HPLC

Article  in  Phytochemistry Reviews · June 2007


DOI: 10.1007/s11101-006-9036-y

CITATIONS READS

73 6,954

2 authors, including:

Mario Jolicoeur
Polytechnique Montréal
133 PUBLICATIONS   2,838 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Degenerative diseases View project

All content following this page was uploaded by Mario Jolicoeur on 25 January 2014.

The user has requested enhancement of the downloaded file.


Phytochem Rev (2007) 6:207–234
DOI 10.1007/s11101-006-9036-y

Analysis of Catharanthus roseus alkaloids by HPLC


Steve Hisiger Æ Mario Jolicoeur

Received: 22 June 2005 / Accepted: 5 October 2006 / Published online: 13 March 2007
 Springer Science+Business Media B.V. 2007

Abstract Catharanthus roseus is a medicinal Examples of spectra obtained using the most
plant from which secondary metabolites used in common detectors are also shown and suggestions
chemotherapy to treat diverse cancers are ex- are made on how to proceed in developing
tracted. The well known high value metabolites efficient separation and identification methods at
vincristine and vinblastine are just 2 of 130 the analytical and semi-preparative scales.
alkaloids that can be found in C. roseus. However,
only few (~11) of this high number of chemical Keywords Semi-preparative HPLC  Reversed-
entities are frequently analyzed and even fewer phase HPLC  Fluorescence  UV  Vincristine 
(~8) are available commercially. For more than Serpentine  Vinblastine
30 years, different analytical techniques have
been developed to isolate and identify C. roseus Abbreviation
metabolites, and then allowing revealing the HPLC high performance liquid
therapeutic potential of C. roseus metabolites. chromatography
Among few approaches, high performance liquid TLC thin layer chromatography
chromatography (HPLC) technique is still widely SPE solid phase extraction
used for the separation and analysis of secondary PDA photo diode array
metabolites such as those from C. roseus. This FW fresh weight
article thus reviews the most recent developments DW dry weight
in HPLC analysis of alkaloids from C. roseus. MS mass spectrometry
Diverse considerations that are crucial to the TIA terpene indole alkaloid
efficiency of secondary metabolites separation and TCA trichloroaceticacid
identification steps, such as biomass manipulation, TEA triethylamine
extraction phase and protocols, HPLC separation TFA trifluoroacetic acid
and analysis protocols are reviewed in details. NMR nuclear magnetic resonance
CD circular dichroism
S. Hisiger  M. Jolicoeur (&)
Canada Research Chair on the Development of
Metabolic Engineering Tools, Bio-P2 Research Unit, Introduction
Department of Chemical Engineering, École
Polytechnique de Montréal, P.O. Box 6079, Centre-
ville Station, Montréal, Québec, Canada H3C 3A7 Catharanthus roseus is known to biosynthesize
e-mail: mario.jolicoeur@polymtl.ca more than 130 alkaloids (van der Heijden et al.

123
208 Phytochem Rev (2007) 6:207–234

2004). Although this rich molecular pool has been plant alkaloids (McCalley 2002; Dräger 2002;
screened for therapeutics since few decades, only Houghton 2002; Molyneux et al. 2002; Stöckigt
five alkaloids from C. roseus (serpentine, ajmal- et al. 2002; Chen et al. 2004). Thin-layer chroma-
icine and the bisindole alkaloids vinblastine, tography (TLC) (De Luca et al. 1986; Asada and
vincristine and 3¢,4¢-anhydrovinblastine) are mar- Shuler 1989; Monforte-Gonzáles et al. 1992) and
keted. Ajmalicine and serpentine are prescribed high-performance TLC (Klyushnichenko et al.
for the treatment of hypertensia, whereas the 1995) were previously suggested for a rapid
bisindoles vinblastine, vincristine and 3¢,4¢-anhy- screening of plants or cell lines. However, these
drovinblastine are used for their antineoplastic methods show a low precision level for the
activity in the treatment of many cancers (see van identification and quantification of alkaloids
der Heijden et al. 2004 for review). These from complex mixtures such as crude plant
secondary metabolites are end-products usually extracts, which contain hundreds of different
found in plant extracts (Fig. 1), and the biosyn- compounds. High-performance liquid chromatog-
thetic pathways leading to these compounds are raphy (HPLC) then represents a powerful capac-
already described in literature (van der Heijden ity of separation of alkaloids and is particularly
et al. 1989, 2004; Facchini 2001; De Luca and adapted for secondary metabolites fingerprinting.
St-Pierre 2000; De Luca and Laflamme 2001). In Automation of HPLC allows high-throughput
vitro cultures of plant cell suspension and of analyses and collection of the different fractions
diverse tissues (such as hairy roots) have been after separation (Stöckigt et al. 2002).
widely studied for the production of these high Among the molecules illustrated in Fig. 1,
value and complex molecules, but with limited current HPLC methods mainly allow for the
commercial success. A major bottleneck is that separation and the identification of ajmalicine,
some of the alkaloids showed to be tissue-specific serpentine, vinblastine, vincristine, vindoline,
and cannot be obtained in undifferentiated cell catharanthine, tabersonine, lochnericine, secolog-
culture. Vincristine and vinblastine require both anin, strictosidine, tryptophan and tryptamine.
aerial and root parts of a plant to be synthesized Reason for this limited list of alkaloids relies on
(De Luca and Laflamme 2001). Horhammericine, the fact that no standards are commercially
tabersonine, löchnericine, 19-hydroxytaberso- available (to our knowledge) for the other alka-
nine, 19-O-Acetyl-hörhammericine and echitove- loids found in C. roseus. Due to their relative low
nine are mostly synthesized in roots (Shanks et al. abundance in plant and cell extracts, unidentified
1998; Laflamme et al. 2001; Rodriguez et al. alkaloids usually appear as weak peaks that are
2003), and are not always detected in suspension ‘‘lost in the forest’’ of a chromatogram; a situation
cells (Kutney et al. 1980; Jolicoeur unpublished which can bias the identification and the quanti-
data). In addition, the natural bisindole alkaloids fication of the compounds of interest. In the
vingramine and methylvingramine have been pathway leading from tabersonine to vindoline,
seen in seeds of C. roseus (Jossang et al. 1998), which occurs in the aerial parts of the plant (De
but their presence in other tissues has not Luca et al. 1988; St-Pierre et al. 1999; De Luca
been demonstrated yet. In the last decades, a and St-Pierre 2000), four intermediates have been
significant research effort has focused on remov- identified as 16-hydroxytabersonine, 16-meth-
ing the metabolic limitations that are encountered oxytabersonine, 16-methoxy-2,3-dihydro-3-hydro-
in in vitro culture as compared to plants (Ver- xy-N-methyltabersonine and deacetylvindoline
poorte et al. 1999; Verpoorte and Memelink 2002; (De Luca and Laflamme 2001). Strictosidine
Hughes et al. 2004). Research in metabolic engi- derivatives as 4,21-dehydrogeissoschizine, cathen-
neering and cell line selection have then signifi- amine, epicathenamine, and stemmadenine and
cantly contributed to the improvement of its derivatives (akuammicine, condylocarpine)
analytical methods, and most of the efforts have leading to catharanthine and tabersonine (Ste-
been placed on chromatographic technologies vens et al. 1993; El-Sayed et al. 2004) can co-elute
which now show to be highly efficient, fast, with compounds of interest (see above). There-
accurate and high-throughput for the analysis of fore, the efficiency of a separation method will

123
Phytochem Rev (2007) 6:207–234 209

tryptamine

NH2
N
strictosidine H

NH
N
H
H OGlc

H
O secologanin
MeOOC
H O

H
OGlc

H
O
MeOOC

4,21-dehydrogeissoschizine cathenamine ajmalicine serpentine

N+ N N N+
N N N N
H H H
H H
H H H
H

H H H H
O O O
MeOOC MeOOC MeOOC MeOOC
OH

stemmadenine tabersonine lochnericine


N N
N O

H H
N
H H N N
H H
MeOOC CH2OH CO2Me CO2Me
N
O

N OH
N H
N
H CO2Me

N OH H
hörhammericine
CO2Me N OH
MeO N
N CH2CH3 H H
H COOMe Me CO2Me CO2Me

19-hydroxytabersonine
catharanthine vindoline

OH
OH
N
N

H
H
N
N N COOMe
N COOMe H
H H
H
OH
OH
MeO N CO2Me
MeO N CO2Me H
H CHO
CO2Me
Me CO2Me

vinblastine vincristine

Fig. 1 Simplified pathways of biosynthesis for the TIAs from Catharanthus roseus. The molecules in bold are those
separated and identified by HPLC. Pathways adapted from Meijer et al. (1993) and Tikhomiroff and Jolicoeur (2002)

123
210 Phytochem Rev (2007) 6:207–234

rely on its capacity to result in pure peaks for the separation methods will rely on the detectors’
known metabolites as well as for the unknowns. sensitivity. This article thus reviews what has been
This constrain of a clear peak separation is not shown to work with C. roseus, from the early steps
obvious and is highly demanding on the analytical of plant cell and tissue harvesting, to the identi-
equipments and protocols required to detect, fication of the secondary metabolites and their
separate, identify and quantify intermediates quantification in extracts.
and end metabolites. On the one hand, the Finally, the emergence of new techniques in
development of a large spectral analytical method combinatorial chemistry for massive synthetic
allowing for the identification of most of C. roseus synthesis and rapid identification of potential
secondary metabolites is required for molecular drugs has led to the development of impressive
screening purposes. On the other hand, with the high-throughput screening (HTS) techniques
recent perspectives of engineering plant cells which can be obviously applied to natural com-
metabolism to enhance productivities in second- pounds (Strege 1999). HTS platforms require
ary metabolites from field and in vitro cultures rapid separation, detection, identification and
(Verpoorte et al. 1999, Verpoorte and Memelink collection of alkaloids from thousands of samples
2002; Rao and Ravishankar 2002; Hughes et al. within a day (van Elswijk and Hirth 2003). From
2004; Capell and Christou 2004), it is crucial to be these platforms, UV, MS-MS and NMR spectra
able to identify and quantify the fluxes controlling libraries have been established ensuring efficient
the secondary metabolic pathways. Quantification recognition of already identified compounds from
of the alkaloids and of the indole and iridoid crude extracts (Abel et al. 2002). Nevertheless,
precursors is then required. The indoles trypto- the generation of massive sets of chromatograms
phan and tryptamine can be analyzed by HPLC from HTS platforms requires efficient automatic
using fluorescence or UV-detection (Dagnino treatment of the data in order to prevent any loss
et al. 1996; Tikhomiroff and Jolicoeur 2002). It of crucial information (Hendriks et al. 2005).
has been established that the biosynthesis of Moreover, in such HTS platforms, HPLC sepa-
secologanin is a limiting step for alkaloids accu- ration technology is a crucial step enabling
mulation in cell culture of C. roseus (Moreno biochemical detection in a continuous flow assay
et al. 1993). Collu et al. (1999) have then devel- to achieve isolation of a lead compound which
oped a HPLC method for analyzing the precur- interacts with a therapeutic target (Ingkaninan
sors geraniol and 10-hydroxygeraniol in an in et al. 2000; van Elswijk and Hirth 2003). Using
vitro enzymatic assay with detection at their such systems, functional and identification assays
maximum UV-absorbance (210 nm). In addition, can be run simultaneously. Despite the emer-
there are now synthetic binary compounds gence of new techniques for the analysis of
derived from natural alkaloids ant that need to alkaloids such as capillary electrophoresis (CE)
be separated and quantified. For instance, vinde- (Chu et al. 1996; Barthe et al. 2002) this review
sine can be derived from vinblastine and vincris- focuses mainly on HPLC methods because it can
tine (Barthe et al. 2002). Coupling vindoline to be scaled-up and used to obtain milligrams of
catharanthine by chemical reaction has led to pure compounds.
synthetic antitumorals anhydrovinblastine, vino-
relbine and vinfluvine (Potier et al. 1975; Kruc-
zinsky and Hill 2001; Fahy et al. 2002). Barthe Detection of Catharanthus roseus alkaloids and
et al. (2002) have developed a HPLC method to precursors
separate these synthetic compounds.
Separation of alkaloids by HPLC analysis is HPLC can be efficiently coupled to different
not only essential for plant and cell line screening, detectors such as Photo Diode Array (PDA),
but also for the design and the validation of fluorescence, ESI-MS, MS-MS, NMR and CD
product recovery and purification processes at the (Bringmann et al. 2002) for precise identification
industrial scale. Therefore, efficiency of the har- and/or quantification of alkaloids. Naaranlathi
vesting procedure as well as the accuracy of et al. (1989) have also proposed electrochemical

123
Phytochem Rev (2007) 6:207–234 211

Table 1 Specific properties of C. roseus’ TIAs and precursors for HPLC separation and detection
Alkaloid Fluorescence (max Ex/Em in nm) Absorption UV (nm) Hydrophilicity (compared to serpentine)

Tryptophan 270/370 218, 278 +


Tryptamine 270/370 218, 278 +
Ajmalicine 270/390 246, 290 +
Serpentine 350/450 252, 308, 370 0
Vinblastine 297/364 214, 266 –
Vincristine 222, 256, 298 –
Vindoline 307/357 214, 254, 306 –
Catharanthine 290/363a 226, 282 –
Tabersonine 222, 302, 326 –
Lochnericine 332 –
Secologanin 238 +
Strictosidine 280/354b 280
a
Renaudin (1985) reported that catharanthine could be detected by fluorescence, but it is 9 times less fluorescent than
ajmalicine and about 380 times less fluorescent than serpentine (at their maximum of fluorescence). Moreover, in the same
article it is mentioned that the sensitivity in a HPLC analysis for catharanthine is better in absorbance than in fluorescence
detection
b
Pennings et al. (1989) found than the strictosidine fluorescence signal intensity is three times smaller than UV absorbance
signal at 280 nm

detection for analyzing cell culture samples. most of the alkaloids. However, 254 nm for
Indeed, there are physical characteristics serpentine and 340 nm for tabersonine-like com-
(Table 1) guiding for efficient HPLC detection pounds will allow higher precision and limit of
of C. roseus secondary metabolites. Since alka- detection, with signal-to-noise ratios of respec-
loids present a structure composed of aromatic tively 4 and 60 times higher than at 218 nm.
rings, UV absorbance is a method of choice for Therefore, since the wavelengths generating max-
their detection. The presence of p-delocalized imal absorbance do not exactly correspond for all
electrons in the alkaloids structure enables auto- alkaloids, a PDA detector having multiple UV
fluorescence phenomena allowing for their effi- channels seems preferable for quantitative studies
cient detection with a reduced baseline back- of alkaloids mixtures. Indeed, setting the detector
ground, especially for the highly fluorescent ones channel to a specific wavelength for a compound
(e.g. serpentine and ajmalicine). Alkaloids are allows enhancing the baseline by suppressing the
also easily ionized by electrospray in positive noise from low-abundant alkaloids or from other
mode, so their detection in ESI (+) MS is recom- UV absorbing interfering molecules and mobile
mended. However, among the detectors used in phase. Moreover, as suggested by Shanks et al.
HPLC analysis, UV and fluorescence are partic- (1998) peak identification may be more reliable
ularly efficient for the quantification of alkaloids, using UV–VIS spectrum obtained by PDA than
and these detectors are easily accessible to the just relying upon retention time. Limits of detec-
large scientific community. tion (LOD) for vindoline and catharanthine were
10 and 0.52 lg ml–1, respectively for the method
Detection upon UV absorbance developed by Naraanlathi et al. (1987) and the
method established by Tikhomiroff and Jolicoeur
As stated above, indole alkaloids can be UV- (2002). Recent developments in PDA detector
detected and identified from their specific UV and separation column technologies have con-
absorbance spectra after their separation as single tributed to significantly decrease the LOD. How-
compound. Typical UV spectra are showed in ever, the limit of detection under UV can be
Fig. 2 (Tikhomiroff and Jolicoeur 2002). If a significantly affected by the presence of other
single wavelength is available, 218 nm then seems compounds at proximity of the peaks of interest
optimal at maximizing the absorbance level for (Chu et al. 1997).

123
212 Phytochem Rev (2007) 6:207–234

75
400

mAU
50

mAU
200
25

0 0
200 220 240 260 280 300 320 340 360 200 220 240 260 280 300 320 340 360
nm nm
Tryptophan Ajmalicine

30 400

mAU
mAU

20
200
10

0 0
200 220 240 260 280 300 320 340 360 200 220 240 260 280 300 320 340 360
nm nm
Secologanin Tryptamine

. 750
600

mAU
500
mAU

400
200 250

0 0
220 240 260 280 300 320 340 360 380 400 220 240 260 280 300 320 34 360 380 400
nm nm
Vincristine Serpentine
.
1000
1500
mAU

1000
500
mAU

500
0 0
220 240 260 280 300 320 340 360 380 400 220 240 260 280 300 320 340 360 380 400
nm nm
Catharanthine Vindoline

600 750
mAU

400 500
mAU

200 250

0 0
220 240 260 280 300 320 340 360 380 400 220 240 260 280 300 320 340 360 380 400
nm nm
Tabersonine Vinblastine

30

20
mAU

10

0
220 240 260 280 300 320 340 360 380 400
nm
Lochnericine

Fig. 2 UV spectra of C. roseus TIAs and precursors. Spectra were recorded using a Beckman-Coulter system Gold 168
PDA detector

Detection upon fluorescence Even if these compounds can either be easily


detected under UV, fluorescence is more sensitive
The corynanthe-type alkaloids such as ajmalicine with a lower limit of detection at the micromolar
and serpentine are fluorescent and can be level. The precursors tryptophan, tryptamine and
detected using a fluorescence detector (Fig. 3). the alkaloid ajmalicine can efficiently be detected

123
Phytochem Rev (2007) 6:207–234 213

Excitation Wavelength (nm)


0 a 0 b proach requires to be validated for each peak
530 530
87.50 325.0 with the identification of the co-eluting com-
490 175.0 650.0 490
262.5 975.0 pounds and by establishing the detection condi-
450 450
350.0
437.5
1300
tions minimizing interference phenomena.
410 1625 410
525.0 1950
370 612.5 2275
370
330 700.0 2600 330
Adequate sample preparation enhances
290 290
analytical steps

c d
Excitation Wavelength (nm)

530
0
500.0
0
100.0
530 An extraction method has to cope with the
490 1000 200.0 490 starting plant material and the differential in the
1500 300.0
450 2000 400.0 450 physico-chemical properties of the molecules to
410 2500 500.0
410 be extracted and of those to be removed; their
3000 600.0
370 3500 700.0 370 respective partition coefficient in the extraction
330 4000 800.0
330 solvent being of the most significant parameters.
290 290 Special cares that have to be considered at each
step for the commnonly used methods are pre-
330
370
410
450
490
530
570
330
370
410
450
490
530
570

sented below.
Emission Wavelength (nm)

Fig. 3 2D-Fluorescence spectra of some C. roseus’TIAs Biomass considerations


and precursors acquired in an aqueous solution pH 5.7
(from Hisiger and Jolicoeur 2005). The fluorescence Secondary metabolites are generally present in a
spectra of tryptophan (a), tryptamine (b), serpentine (c) wide range of concentrations that may even vary
and ajmalicine (d) were recorded with a multichannel
fluorescence sensor (Delta Light and Optics, Lyngby, from one lot or batch to another. Therefore, large
Denmark) using 12.5 mg l–1 standard solutions amount of biomass may be required to have access
to a large spectrum of compounds each at an
at Ex 270/Em 370 nm, despite the fact that the adequate concentration. Usual amounts of bio-
maximum excitation wavelength for ajmalicine is mass sampled from in vitro cultures and whole
Ex 290 nm, whereas it is Ex 270 nm for both plant culture are presented in Tables 2 and 3,
precursors. Limits of detection for tryptophan respectively. Freeze-drying of the biomass enables
and tryptamine were lower to 0.01 lg ml–1 (Tik- to crush the cells, to perform extraction steps on
homiroff and Jolicoeur 2002). Renaudin (1985) concentrated samples which decrease the volume
reported that the limit of detection can be to be processed without denaturing alkaloids.
decreased by a factor of six for ajmalicine and From in vitro plant cell and tissue cultures,
vindoline and even by a factor of 50 for serpen- 50 mg DW have been reported to be the lower
tine by using fluorescence detection instead of biomass limit for the efficient quantification of
traditional UV detection. Combining the use of most of the known alkaloids. This sample size
fluorescence and absorbance detectors, molecules represents 0.5–1 g FW and thus up to 3 ml of a
that co-elute but which show different physical plant cell suspension in liquid culture. This is seen
characteristics can still be quantified by selecting as a minimum and may not always allow for the
appropriate wavelengths. For instance, quantifi- detection of all alkaloids, even for the known
cation of serpentine performed at 306 nm was ones. Sample concentration can be increased in
possible while co-eluting with an unidentified different ways such as evaporation of the extrac-
compound (Tikhomiroff and Jolicoeur 2002). tion phase and re-suspension into a smaller
Renaudin (1985) showed that co-elution of ser- volume of solvent, or by using solid phase extrac-
pentine with vinblastine has no effect on serpen- tion as for aqueous solution extraction (see
tine response factor since no quenching occurs; below), and then modulating the elution volume
they can then be differentiated from their respec- (Renaudin 1985; Lee-Parsons and Shuler 2002). In
tive fluorescence spectra. Nevertheless, this ap- most cases, less than few micrograms of pure

123
214 Phytochem Rev (2007) 6:207–234

Table 2 Protocols for alkaloids extraction from C. roseusroots or hairy root


Authors Biomass Exraction phase Procedure

Tikhomiroff and 200 mg FW Methanol – Lyophilization


Jolicoeur (2002) hairy roots – Crushing of dry material in a tissue grinder
– Extraction with 1 ml for 1 h in a sonicating bath
Morgan et al. (2000) 100 mg DW Methanol – Lyophilization
– Crushing of dry material with a mortar
– Extraction with 45 ml for 5 h in a sonicating bath
– Evaporation of the mobile phase
Sim et al. (1994) 50 mg DW Methanol then – Extraction with 3 · 10 ml of MeOH for 30 min
ethyl acetate in a sonicating bath at 50C
– Evaporation of the methanol
– Resupension in 20 ml 0.1 N HCl
– Extraction with 20 ml Ethyl acetate
– Adjusting to pH 10
– Extraction three times with ethyl acetate
– Evaporation
– Resupension in methanol
Bhadra et al. (1993) 200 mg DW Methanol – Lyophilization
– Extraction with 80 ml for 3 h in a Soxhlet apparatus
– Evaporation and dilution in a 5 mM (NH4)2HPO4 solution
– Fractionation on a 300 mg-C18 cartridge with three successive
elutions with a mixture of MeOH: (NH4)2HPO4,
60:40, 95:5 and 100:0 (v:v)
Hughes et al. (2004) 50 mg DW Methanol – Lyophilization
– Extraction with 10 ml of methanol for 1 h in a sonicating bath
– Centrifugation for 15 min at 1300g, 15C
– Repetition of the extraction step and combination of the both
extracts
– Concentration under vacuum to 2 ml

alkaloids can be obtained from a 50 mg DW Uniyal et al. 2001; Singh et al. 2000; from cell
sample. However, milligrams of pure alkaloids are culture: Dagnino et al. 1996; from hairy roots:
required for obtaining standards that are required Tikhomiroff and Jolicoeur 2002). Increasing the
for the identification of unknowns or for assessing volume of the extraction phase over the biomass
biological activity of the molecules, and thus kg of amount increases obviously the extraction capac-
cells and tissues will then be needed. ity. However, using large volume will dilute the
compounds and it will be necessary to add
Selection of an extraction phase concentration steps prior to analysis. Performing
a single step extraction on 20 mg DW of hairy
Two major extracting phases are suggested in roots with 1 ml methanol, Tikhomiroff and Jolic-
literature for the extraction of alkaloids, such as oeur (2002) were able to extract virtually 100% of
methanol and ethyl acetate, but ethanol (Balagué the secologanine and ajmalicine contents, but the
and Wilson 1982; Naaranlahti et al. 1987; Moreno extraction yields were significantly lower for the
et al. 1993, Singh et al. 2000; Uniyal et al. 2001), other alkaloids. However, several extraction steps
phosphoric acid (Blom et al. 1991), acetic acid with fresh solvent are usually required for a better
(Renaudin 1985) and acetone for secologanin recovery. Besides classical extraction procedures,
(Contin et al. 1998) were also used. Different Choi et al. (2002) tested the use of supercritical
ratios of solvent volume-to-biomass have been fluid (mix of CO2, methanol and triethylamine)
tested (Tables 2, 3 and 4). In general, typical ratio for the recovery of vinblastine and vincristine
of 0.2–0.6 ml solvent mg–1 DW is used. However, from C. roseus leaves as an alternative to pre-
some authors prefer to perform their extraction at purification steps. However, the extraction yield
lower ratio of 0.02–0.1 ml mg–1 DW (from leaves: was at best 76.6% of that using methanol only.

123
Phytochem Rev (2007) 6:207–234 215

Table 3 Protocols for alkaloids extraction from C. roseus leaves


Authors Biomass Extraction Procedure
phase

Uniyal et al. 5 g of oven-dried Ethanol – Oven-drying of the leaves for 48 h at 60C


(2001) tissue – Extraction (3 · 30 ml) overnight at room temperature
– Filtration and concentration in vacuo at 4C
– Dissolution in ethanol and dilution in 10 ml water
and 10 ml 3 mM HCl
– Washing with hexane (3 · 30 ml)
– pH 8.5 adjusting of the aqueous phase and cool down at 10C
– Extraction with chloroform (3 · 30 ml)
– Washing with water
– Evaporation of the chloroform phase
– Resuspension in 1 ml of chloroform
– Fractionation on SPE cartridge and elution with a
chloroform:methanol 90:10 solution
– Evaporation then resuspension in Methanol
Singh et al. 5 g DW Ethanol – Extraction with 3 · 30 ml ethanol (12 h each at room temp.)
(2000) – Filtration and concentration to 10 ml
– Add 10 ml of water and 10 ml of 3% HCl
– Washing three times with hexane
– Alkalinization of the aqueous phase to pH 8.5
– Extraction with chloroform (3 · 30 ml)
– Washing with water
– Concentration in vaccu-oven
– Dissolution in 10 ml methanol

Selection of the extraction phase is crucial and pre-cleaning purposes removing contaminating
depends on the solubility of the alkaloids. Acidic compounds. For instance, TCA and acetone are
extractive solvents (pH = 1.5) are commonly used well known to cause protein precipitation,
and it seems to increase the stability and the whereas alkaloids remain in solution, and TFA
solubility of the alkaloids (Hallard et al. unpub- seems to cause precipitation of chlorophyll (Hal-
lished results; see caption of Fig. 4 for the lard et al. unpublished results; see caption of
complete reference). Solvent phase selection is Fig. 4 for the complete reference).
also dependent on the nature of the analysis to be
performed. For instance, using methanol as the Sample preparation
extraction phase and then running a HPLC
method with phosphate buffer should be avoided Cell membrane and debris as well as precipitated
because methanol can precipitate phosphate salts macromolecules have to be removed from the
in different HPLC parts (e.g. tubings, column and solvent after sample extraction. Liquid sample
pre-column, detectors, etc.). In addition, when clarification is usually achieved by centrifugation
further molecular characterization has to be at high speed (5 min, 15,000g) and/or by filtration
performed using MS detector, non-volatile sol- (see Table 3 for details). The liquid sample has to
vents (especially phosphoric acid) should be be filtered through a submicron filter. A nylon or
proscribed in order to prevent any damage to PTFE membrane is adequate for solvents sug-
the MS detector. For MS analysis, TFA should be gested in literature (e.g. methanol or acetonitrile).
used with precaution because high TFA concen- This step is essential to extend the HPLC column
tration (0.1% w/v) causes ion-suppression in MS life-time as well as the global performance of the
and peak tailing can then be observed (Gustavs- HPLC, including that of the injector. Even when
son et al. 2001; Annesley 2003). Finally, one may using SPE columns (Solid Phase Extraction),
require performing several successive extractions filtration/centrifugation is a pre-requisite step
using the same solvent or even different ones for before cartridge loading.

123
216 Phytochem Rev (2007) 6:207–234

Table 4 Protocols for alkaloids extraction from cell suspension of C. roseus


Authors Biomass Extraction phase Procedure

Dagnino et al. (1996) 50 mg Dichloromethane – Freeze-drying of the biomass


(apolar alkaloids) DW – Wetting with 0.5 ml of water
– Extraction with 5 ml of dichloromethane (repeat one more
time)
– Drying and resuspension in acetonitrile
Dagnino et al. (1996) 50 mg Methanol – Extraction with 5 ml of methanol of the dichloromethane
(alkaloids precursors) DW extracted biomass (repeat one more time)
– Drying and resupension in 0.5 ml 1 M phosphoric acid
Renaudin (1985) 1–3 g Acetic acid – Freezing and thawing of the fresh cells
FW – Extraction four times with 15 ml 0.01% (v/v) acetic acid
(pH 4.0)
– Centrifugation 4200g for 5 min
– Adjusting the pH between 7.3 and 7.5 with 1 M NaOH
– Loading a SPE C18 cartridge
– Rinsing with 60 ml methanol–25 mM ammonium dihydrogen
phosphate (pH 7.3) (10/90, v/v)
– Elution with 2 ml methanol–25 mM ammonium dihydrogen
phosphate (pH 4.7) (85/15, v/v)
Lee-Parsons et al. (2004) 250 mg Methanol – Lyophilization
DW – Extraction with 25 ml methanol (one more time) for 3 h
– Evaporation to dryness
– Dissolution in an aqueous solution 10% acetonitrile + 0.1%
TFA
– Loading on SPE C18
– Elution with 100% acetonitrile plus 0.1% TFA
– Evaporation and dissolution in aqueous 20% acetonitrile
plus 0.1% TFA
Monforte-Gonzáles et al. 1 g DW Methanol then – Lyophilization
(1992) ethyl acetate – Extraction with 50 ml methanol for 2 min in a Polytron
homogenizer
– Incubation for 2 h at 55C
– Filtration
– Evaporation and dissolution in 15 ml H2SO4
– Washing with 15 ml ethyl acetate (three times)
– Adjusting the pH to 9.5
– Extracting with 20 ml ethyl acetate (three times)
– Evaporation and dissolution in 1 ml methanol

Extraction protocols then extracted using selected organic solvent.


Solvent volume and the number of extraction
Extraction of alkaloids from plant tissues or cells, steps need to be optimized in order to maximize
from culture medium and from adsorbent resins is extraction yield. Since studies generally focus on
well documented. Commonly used techniques for a particular type of biomass (e.g. cell suspension
TIAs and precursors are summarized in the or hairy roots or plants), extraction protocols are
following. generally biomass type specific. However, differ-
ent tissues can be extracted using a unique
From biomass protocol, as shown by Favali et al. (2004) ana-
lyzing leaves, stems and roots from plants
Alkaloids extraction can be performed starting infected with phytoplasmas. In these works,
with hard tissues such as plant leaves, stems, tissues were extracted in a Soxtec system using
roots (including hairy roots) and cell suspension. methanol at 190C for 2 h. However, the result-
After harvesting, plant material is frozen in ing extraction efficiency may be then lower
liquid nitrogen, ground at cold, freeze-dried and overall.

123
Phytochem Rev (2007) 6:207–234 217

with a strong solvent such as 100% methanol or


100% acetonitrile is then recommended. Some
previous SPE methods used diatomite or cation-
exchange as solid phase (see the nice review from
van der Heijden et al. 1989). Now-a-days, octade-
cylsilyl is preferred for alkaloids preparation (Lee
and Shuler 2000; Renaudin 1985; Uniyal et al.
2001; Bhadra et al. 1993; McCalley 2002).

From adsorbent phases

Fig. 4 UV chromatogram registered at 254 nm of a


Many research groups refer to the use of a solid
standard mixture of C. roseus alkaloids using the method extraction phase in situ in the culture medium of
developed by Hallard D, Vera Rocha RA, Sajjadi SE, van C. roseus suspension cells (Payne et al. 1988;
der Heijden R and Verpoorte R (unpublished results). Asada and Shuler 1989; Lee-Parsons and Shuler
Reproduced with the kind permission of Dr Verpoorte. 1,
tryptamine; 2, perivine; 3, vindolinine; 4, yohimbine; 5,
2002; Wong et al. 2004) and hairy roots (Sim et al.
strictosidine; 6, ajmalicine; 7, serpentine; 8, catharanthine; 1994). The most documented use of a solid
9, vincristine; 10, vindoline; 11, vinblastine, 12, anhydro- extraction phase in plant cell culture is the
vinblastine. The chromatographic conditions are described Amberlite XAD-7 resins (but also XAD-4 and
in details in Table 7
others are used). This resin is made of a neutral
polymeric adsorbent that has shown a fast
adsorption rate and a weak selectivity among
From a liquid medium alkaloids. XAD-7 resins were successfully em-
ployed to enhance the catharanthine and ajmal-
Some authors analyze directly the alkaloids con- icine production levels in cell culture. Liquid
tent of a culture medium without any preliminary extraction phases have also been used in situ in
steps (Dagnino et al. 1995; Whitmer et al. 2002; two-liquid-phase bioreactors, such as silicon oil
El-Sayed et al. 2004). However, the level in (Byun and Pedersen 1994; Tikhomiroff et al.
alkaloids that are secreted in the medium is 2002) and trycaprylin (Collins-Pavao and Chin
generally too low to allow for a good detection 1996). However, liquid extraction phases usually
upon direct HPLC injection. It then becomes show high selectivity for some of the alkaloids.
essential to concentrate TIAs in the liquid sample. Finally, after separation from the medium and
Several methods can be used such as evaporation the cells, alkaloids are extracted from adsorbents
at low temperature (e.g. lyophilization) and resins using methanol (Asada and Shuler 1989;
liquid–liquid extraction (Asada and Shuler 1989; Wong et al. 2004) or methanol:HCl (95:5, v:v).
Zhao et al. 2001; Satdive et al. 2003). The quicker Similar protocols can be applied extracting alka-
and most efficient method is a solid phase extrac- loids from liquid extraction phases (Tikhomiroff
tion using commercial cartridges with C18 packing et al. 2002).
(Renaudin 1985; Hisiger and Jolicoeur 2005). In
addition to sample concentration, the use of SPE Estimation of the extraction yield
cartridge allows to ‘‘clean’’ a sample. Based of the
strength of the solvent used for the elution step, There is no universal method which can be
SPE cartridge can help to fractionate the sample. applied for the extraction of secondary metabo-
It then becomes possible to generate different lites from C. roseus because TIAs have different
fractions, based on the affinity of the compound hydrophobicity levels. The selection of the extrac-
for the stationary phase, as for solid-chromatog- tion phase is then determinant to reach high
raphy techniques (Bhadra et al. 1993; Renaudin extraction yields and every step should be care-
1984). If the aim is to recover all the alkaloids and fully optimized. A typical way in approximating
precursors within a unique liquid phase, elution the extraction efficiency consists in spiking the

123
218 Phytochem Rev (2007) 6:207–234

sample with a known amount of standard material Table 5 Some of commercially available C. roseus alka-
as soon as at the first step of the extraction loids
process; while crushing the tissues. However, this Compound Supplier
method only gives an approximation because
there is a bias caused by the interaction of the Vinblastine SIGMA (USA)
Spectrum chemicals (USA)
spiked alkaloids with the intracellular matrix, a Vinkem Labs (India)
phenomena which can hardly be characterized. Vincristine SIGMA (USA)
Indeed, it has been suggested than some alkaloids Eli Lilly (USA)
can form complexes with phenolics (Renaudin Synnad Chemical Co (China)
Southern Herbals Ltd (India)
et al. 1982; Renaudin 1989). Tikhomiroff and Catharanthine Dayang Chemicals (China)
Jolicoeur (2002) have reported low extraction Chemos GmBH (Germany)
yields (<60%) when using a single methanol Kang’ai biological Products (China)
extraction step for recovering intracellular ser- ZYF Pharm Chemical (China)
Serpentine Apin chemicals (UK)
pentine, vincristine, vindoline, cathranthine, Ajmalicine Extrasynthèse (France)
tabersonine and tryptophan from plant cells Yohimbine Extrasynthèse (France)
whereas the yield was more than 85% for Vindoline Kang’ai biological Products (China)
vinblastine, tryptamine and ajmalicine. Interest- Tryptamine SIGMA (USA)
ingly, Naaranlathi et al. (1987) found a 100% Note: It is not possible to screen for all the suppliers
extraction yield when spiking their leaves samples around the world for C. roseus molecules, but this table
may be useful to find quickly a supplier for a desired
with vindoline and catharanthine after extraction standard
with ethanol. Multiple successive extractions of
plant cells using acetone revealed an efficiency of
recovery for pure secologanin of only 7.5% the major problem here relies on the fact that
(Contin et al. 1998). Matrix effect can also hap- alkaloids and their precursors respond in absor-
pen during HPLC quantification. The use of an bance or fluorescence at different levels. For
internal standard, added to the sample, is thus instance, tryptamine is 3 times more fluorescent
suggested to asses for matrix effect, extraction than tryptophan and 6 times more than ajmalicine
efficiency and injection accuracy. It is then nec- at 270/370 (Fig. 3). The alkaloids concentration
essary to choose a molecule which is absolutely distribution in plant, plant cells and tissues is also
not present in the liquid sample, exhibits the same not similar neither constant with time, so it
behavior as the target compound(s) without co- becomes highly hazardous to establish a calibra-
eluting which would interfere with the HPLC tion between the signal in absorbance or fluores-
analysis. 5-Methoxytryptamine has been used as cence and the total alkaloid content of a sample.
internal standard by Naaranlathi et al. (1987) This method is thus highly pseudo-quantitative
although the use of stable isotopic analogues and is thought to be mostly useful for a quick pre-
(deuterium labeled) can be even more efficient screening of plant or cell lines. In addition, the
when an MS detector is available (Auriola et al. samples have to be thoroughly cleaned (see
1991). Sample preparation section) to avoid any inter-
ference from the other metabolites that absorb in
Pseudo-quantification methods UV or do fluoresce.
In the case of C. roseus, only few of the
In the worst case when no standards are available, required standards can be found commercially
one can estimate the total alkaloid content of a (Table 5). Therefore, it is difficult to obtain most
sample selecting a wavelength at which most of of the standards. It is then feasible again to
the alkaloids show a maximal response in absor- proceed by approximation performing external
bance or fluorescence. According to Monforte- calibration using an available and quantifiable
González et al. (1992), total alkaloids can be alkaloid as the reference. Indeed, hörhammeri-
monitored by measuring the absorbance at cine and lochnericine can be quantified, based on
280 nm using a spectrophotometer. However, the response factor of tabersonine as they possess

123
Phytochem Rev (2007) 6:207–234 219

similar UV spectra and molecular extinction


Elution gradient coefficients (Rijhwani and Shanks 1998; Morgan

1.5 ml min–1

1.2 ml min–1
and flow-rate

1 ml min–1
et al. 2000). In a similar way, coronaridine was
Isocratic,

Isocratic,

Isocratic,
quantified upon the response factor of catharan-
thine (Morgan and Shanks 2002). Compounds
which have not the same response factor could
nevertheless be quantified, proceeding as follow.
Ex270/Em370
Fluorescence

It is then important to consider the molecular


Detection

extinction coefficient ratio for both molecules


independently quantified, in the mobile phase at
PDA

PDA their respective retention time, and for the same


NaH2PO4 in methanol:water molar amount loaded on the HPLC column.
Thus, this suggests an initial calibration step using
1% formic acid–acetonitrile–
Acetonitrile:100 mM H3PO4

external standards to assess for analytical bias


LiChrosorb RP-8 Select B, 7 lm 7 mM SDS and 25 mM

such as the matrix effect which can play on co-


(pH 6.2) (68:32, v:v)
trichloroacetic acid
(pH 2) (15:85, v:v)

(100:10:0.25 v:v:w)

eluting alkaloids.
Mobile Phase

Designing, selecting and transposing HPLC


methods

Numerous HPLC methods have been developed


for the analysis of alkaloids and precursors
5 lm 250 · 4 mm (Merck)
LiChrospher 60 RP Select B

content in C. roseus extracts (Tables 6 and 7).


250 · 4.6 mm (Agilent)

In spite of several studies using C8 columns


250 · 4 mm (Merck)
Zorbax XDB C185 lm,

(Gorog et al. 1977; Pennings et al. 1989) and


Phenyl columns (van der Heijden et al. 1987),
there is now a consensus for reverse phase
This alkaloid was quantified using the methods for TIAs precursors

chromatography using C18 columns.


Table 6 HPLC methods for the analysis of the C. roseus precursors
Column

HPLC methods for the analysis of TIAs

TIAs are generally more hydrophobic than their


time (min)
Compounds focused Retention

indole and iridoid precursors. It is then preferable


to develop a set of two HPLC separation meth-
3.2
3.5
7.2

5.5

5.6
12.2

13.5
22.5
25.5
30.5

ods, one for hydrophilic (precursors) and one for


hydrophobic (TIAs) molecules. A more polar
enzymatic assay)
tryptamine (from

elution is then performed when analyzing precur-


sors as compared to alkaloids. The different
Loganic acid,

Strictosidine,
Tryptophan,

secologanin,

secologanin,
tryptophan,
tryptamine,

ajmalicinea

tryptamine

methods that have been developed are presented


loganin,

in Table 7. It can be seen that some methods are


highly similar and may only differ because of
slight differences in equipment performance or
because of specific lab research goals. However,
Jolicoeur (2002)
Tikhomiroff and

one can obviously start developing a separation


Pennings et al.
Dagnino et al.

method from published works. Bhadra and


Shanks (1997) modified a method previously
(1995)

(1989)
Authors

reported by Bhadra et al. (1993) changing the


C18 lBondapak 100 · 8 mm column (Waters) for
a

123
Table 7 HPLC methods for the analysis of alkaloids from C. roseus
220

Authors Compounds Retention Column Mobile Phase Detection Elution gradient


focussed time (min)

123
Moreno- Ajmalicine, 21.8 Ultrasphere ODS, Acetonitrile:10 mM UV Isocratic, 1.5 ml min–1
Valenzuela yohimbine 5.9 4 lm 250 · 4.6 mm (NH4)2HPO4 280 nm
et al. (1998) (Beckman) (43:57, v:v)
Renaudin Ajmalicine, 14 lBondapack C18 Methanol:5 mM UV Isocratic, 1 ml min–1
(1984) vindoline, 9.5 10 lm (NH4)2HPO4 254 nm
catharanthine, 16 300 · 3.9 mm (pH 7.3) (67:33, v:v)
tetrahydroalstonine 22 (Waters)
Renaudin Ajmalicine, 12 lBondapack C18 Methanol:5 mM UV Isocratic, 1 ml min–1
(1984) catharanthine, 13 10 lm (NH4)2HPO4 254 nm
vinblastine, 16 300 · 3.9 mm (pH 7.3)
serpentine 19 (Waters) (71:29, v:v)
Bhadra et al. Ajmalicine, 12 lBondapack C18 Methanol:5 mM UV Isocratic, 0.8 ml min–1
(1993) serpentine, 59.2 10 lm (NH4)2HPO4 254 nm
catharanthine, 14.5 100 · 8 mm (pH not mentioned)
vindoline, 8.5 (Waters) (67:33, v:v)
vinblastine nd
Rijhwani Ajmalicine, N/A C18 bondclone RP Methanol:acetonitrile: PDA Isocratic, 1 ml min–1
and Shanks serpentine, 10 lm 5 mM (NH4)2HPO4
(1998) tabersonine, 300 · 3.9 mm (32:32:36, v:v:v)
hörhammericine, (Phenomenex)
lochnericine
Lee and Ajmalicine, 19.67 lBondapack C18 Acetonitrile: 0.1% UV Isocratic, 1 ml min–1
Shuler catharanthine, 21.60 10 lm (w:v) TFA in water 254 nm
(2000) serpentine 23.14 300 · 3.9 mm (22:78, v:v)
(Waters)
El-Sayed Stemmadenine, 7.2 Vydac C18 RP Acetonitrile:0.1% (w:v) PDA Isocratic, 1 ml min–1
et al. catharanthine, 21.7 250 · 4.6 mm TFA in water
(2004) tabersonine 23.5 (Vydac) (21:79, v:v)
Sim et al. Catharanthine, N/A lBondapack Methanol:acetonitrile: UV Isocratic, 1 ml min–1
(1994) ajmalicine C18 10 lm 5 mM 298 nm
300 · 3.9 mm (NH4)2HPO4(pH 7.3)
(Waters) (30:40:30, v:v:v)
Phytochem Rev (2007) 6:207–234
Table 7 continued
Authors Compounds Retention Column Mobile Phase Detection Elution gradient
focussed time (min)

Uniyal et al. Vincristine, 14.62 Symmetry C18 5 lm Solvent A (pH 7.0): PDA 120
(2001) catharanthine, 15.92 150 · 4 mm Methanol:acetonitrile: 100
vindoline, 19.6 (Waters) 5 mM CH3COONH4:
vinblastine 18.02 triethylamine (13:32:55:0.2, 80
by volume); 60

%B
Solvent B (pH 7.0):
Methanol:acetonitrile:5 mM 40
Phytochem Rev (2007) 6:207–234

CH3COONH4:triethylamine 20
(19:46:35:0.2, by volume)
0
0 10 20 30 40
Time (min)
0-15 min : 0.4 mL min-1
15-20 min : 0.5 mL min-1
20-25 min : 0.7 mL min-1
25-30 min : 0.9 mL min-1
30-35 min : 1.2 mL min-1

Chu et al. Vincristine, 14.8 Hypersil ODS, 5 lm Solvent A: 25 mM ESI-MS 80


(1997) vinblastine 21.3 200 · 4.6 mm CH3COONH4 in water; 60
(Agilent) Solvent B: 25 mM
CH3COONH4in methanol 40
%B

20

0
0 10 20 30
Time (min)
-1
1 mL min

Choi et al. Vinblastine, 18 Zorbax Bonus RP-18, Solvent A: 20 mM ESI-MS 80


(2002) vincristine 21.5 5 lm 150 · 2.1 mm CH3COONH4; Solvent B:
(Agilent) Acetonitrile 60

40
%B

20

0
0 15 30 45
Time (min)
0.2 mL min-1
221

123
222

Table 7 continued
Authors Compounds Retention Column Mobile Phase Detection Elution gradient

123
focussed time (min)

Naaranlahti Vindoline, 18 Spheri-5 RP-18, 5 lm Solvent A: 0.2% PDA 80


et al. catharanthine, 20 220 · 4.6 mm TEA in 25 mM
(1987) serpentine, 13 (Brownlee) CH3COONH4; 60
vinblastine, 24.5 Solvent B: 0.2%
vincristine, 21.5 TEA in a mix of 40

%B
ajmalicine 21 methanol:acetonitrile
(29:71, v:v)
20

0
0 10 20 30
Time (min)
0-17 min : 1 mL min1
17-30 min :1.5 mL min1

Zhao et al. Serpentine, N/A Nucleosil 5 C18 5 lm Methanol:acetonitrile: PDA Isocratic, 1 ml min–1
(2001) ajmalicine, 250 · 4.6 mm 25 mM CH3COONH4:
catharanthine (Agilent) TEA (15:40:45:0.1,
by volume)
St-Pierre and 16-methoxy- 30.5 Nova-Pak C18, 4 lm Methanol:water:TEA PDA Isocratic,
De Luca tabersonine 300 · 3.9 mm (75 :25 :0.1, v:v:v) 0.6 ml min–1
(1995) (from (Millipore)
enzymatic
assay)
Zhou et al. Vindoline, N/A Direct flow injection Methanol ESI-MS/ Injection flow-rate
(2005) vindolidine, MS of the sample 3 ll min–1
vincristine,
vinblastine
Zhou et al. 19S-vindolinine, 13.9 Zorbax Eclipse XDB Solvent A: UV 100
(2005) vindolinine, 16.6 C8, 5 lm, Water, 0.1% TEA; 280 nm,
80
vindoline, 21.6 150 · 4.6 mm Solvent B: ESI-MS/
vindolidine, 25.1 (Agilent) Methanol MS 60
catharanthine 27.1
%B

vincristine, 28.2 40

vinblastine 36.5 20

0
0 20 40 60
Time (min)
0.8 mL min-1
Phytochem Rev (2007) 6:207–234
Table 7 continued
Authors Compounds Retention Column Mobile Phase Detection Elution gradient
focussed time (min)

Aerts et al. Catharanthine, N/A Hypersil ODS, Solvent A: Water, PDA 80


(1994) vindoline, 5 lm 200 · 2.1 mm 0.1% TEA;
tabersonine (Agilent) Solvent B: 60
Acetonitrile,
0.1% TEA 40
Phytochem Rev (2007) 6:207–234

%B
20

0
0 10 20 30
Time (min)

Barthe et al. Vindoline, 6.0 XTerra RP-18, 5 lm Acetonitrile:water:boric UV Isocratic, 1 ml min–1


(2002) viorelbine, 14.6 250 · 4.6 mm acid (pH 10) (55:45:3.1, 214 nm
anhydrovinblastine, 20.8 (Waters) v :v :w)
vinflunine, 10.4
catharanthine 8.8
Auriola et al. Tryptamine, 2 lBondapak C18, Acetonitrile:0.1 M UV Isocratic, 1 ml min–1
(1989) ajmalicine, 5.5 10 lm CH3COONH4 280 nm,
serpentine, 8 300 · 3.9 mm (pH 7.2) (49:51, v:v) TSP-MS
catharanthine, 10
tabersonine 18
Toivonen Ajmalicine, N/A Hypersil ODS, 5 lm Solvent A: 14 mM TEA PDA 60
et al. catharanthine, 100 · 2.1 mm in CH3COONH4;
(1991) tabersonine (Agilent) Solvent B: 14 mM
TEA in Methanol: 40

acetonitrile (50:50, v:v)


%B

20

0
0 10 20 30 40
Time (min)
0.4 mL min-1
223

123
224

Table 7 continued
Authors Compounds Retention Column Mobile Phase Detection Elution gradient

123
focussed time (min)

Tikhomiroff Catharanthine, 14.5 Zorbax XDB C18 Solvent A: 5 mM PDA 80


and serpentine, 7.8 5 lm KH2PO4 (pH 6);
60
Jolicoeur vindoline, 13.5 250 · 4.6 mm Solvent B:
(2002) vincristine, 13.1 (Agilent) Acetonitrile 40

%B
vinblastine, 15.2
tabersonine 20.8 20

0
0 10 20 30
Time (min)
2 mL min-1

Singh et al. Catharanthine, 12.32 lBondapack C18, Acetonitrile:0.1 M PDA Isocratic, 0.6 ml min–1
(2000) vindoline, 13.54 10 lm Phosphate buffer:glacial
vincristine, 10.15 300 · 3.9 mm aetic acid (pH 4.14)
vinblastine 11.34 (Waters) (38:62:0.3, v :v :v)
Lee-Parsons Tryptamine 5 Luna C18 5lm Solvent A: Water, UV 25

et al. ajmalicine, 34 150 · 4.6 mm Solvent B: 254 nm 20


(2004) serpentine 37 (Phenomenex) Acetonitrile,
15
Solvent C:
%B

Formic acid (kept at 10

0.1% of the total elution 5


volume)
0
0 10 20 30 40 50
Time (min)
1 mL min-1

Schröder Tabersonine, 26.7 Nucleosil C18 Solvent A: 0.2% NaOH PDA 80


et al. 16-hydroxy- 5 lm 200 · 4 mm in water,
60
(1999) tabersonine, (Agilent) Solvent B:
16-methoxy- Acetonitrile 40
%B

tabersonine
20
13.0
26.2 0
0 10 20 30
Time (min)
1 mL min-1
Phytochem Rev (2007) 6:207–234
Table 7 continued
Authors Compounds Retention Column Mobile Phase Detection Elution gradient
focussed time (min)

Hallard et al., Tryptamine, 3.5 ‘‘Low TFA’’ C18 Solvent A: UV 254 nm, 15
(unpublished) secologanin, 4.0 5 lm TFA:water:acetonitrile ESI-MS
perivine, 4.2 250 · 4.6 mm (0.01:79:21); 10

%B
vindolinine, 4.9 (Vydac) Solvent B: 5
yohimbine, 5.2 TFA:water:acetonitrile
strictosidine, 5.9 (0.01:5:95) 0
ajmalicine, 8.1 0 10 20 30 40
Time (min)
Phytochem Rev (2007) 6:207–234

serpentine, 8.6
alstonine, 9.2
1 ml min-1
catharanthine, 9.3
vincristine, 12.5
tabersonine, 13.5
vindoline, 16.3
cathovaline, 18.8
vinblastine, 21.1
anhydro- 26.4
vinblastine
225

123
226 Phytochem Rev (2007) 6:207–234

a C18Bondclone, 10 lm, 300 · 3.8 mm column observed that an old column (hopefully after
(Phenomenex), and using a gradient flow running few thousands of samples) offers a
mode with a mixture of methanol and 5 mM weaker retention of alkaloids with low pKa (e.g.
(NH4)2HPO4 (pH 7.3) (58:42, v:v). Using the ajmalicine) but a stronger retention of com-
modified method, pure peaks were obtained for pounds with high pKa (as tryptamine and ser-
ajmalicine, serpentine and tabersonine but cath- pentine) than a new column (Renaudin 1984).
aranthine could not be quantified due to UV This phenomenon is thought to be due to the
interfering compounds. With the same Bondclone degradation of the hydrophobic stationary phase
column, quantification of ajmalicine, serpentine, resulting from the uncapping of the silanol groups
tabersonine, hörhammericine and lochnericine following the disruption of the C18 alkyl chains.
was successful using an isocratic elution at The presence of amine modifier agents in the
1 ml min–1 (Table 7). Alkaloids metabolism in mobile phase modulates the interaction of the
wounded seedlings was studied by Vazquez-Flota stationary phase with basic analytes (e.g. alka-
et al. (2004) using the method previously devel- loids) and can thus change the elution sequence.
oped by Aerts et al. (1994) in a study on the effect It then becomes tedious to compare the methods.
of methyl jasmonate vapor on C. roseus seedlings. Competitive amines can be used to enhance
The method developed by Tikhomiroff and peaks shape at neutral pH and reducing peak
Jolicoeur (2002) for screening secondary metab- tailing (Nawrocki 1997; McCalley 2002). Mobile
olites in hairy roots, was recently used for the phase pH is an important parameter and acidic
analysis of the accumulation of alkaloids in pH (pH 3–4) enables better peak shape for basic
different plant cultivars (Dutta et al. 2005). alkaloids. At low pH, most of silanol groups are
There are multiple approaches when develop- undissociated and ion-exchange interactions are
ing a HPLC separation method and the following then limited, even if alkaloids are protonated
basic considerations have to be considered. In the (McCalley 2002). Alkaline pH (pH 10) was only
case of TIAs analysis, there is no consensus on tested by Barthe et al. (2002), to our knowledge.
mobile phase selection. An effective separation Multiple columns can be found commercially if
can be achieved using a saline buffer with one chooses to work at extreme alkaline pH but it
potassium phosphate or ammonium acetate as should be noted that past-generation columns
aqueous phase, and methanol or acetonitrile as (e.g. C8, Phenyl and C18) cannot be used at high
organic phase (Bhadra et al. 1993; Moreno-Val- pH because the destruction of the stationary
enzuela et al. 1998; Choi et al. 2002; Tikhomiroff phase may be induced. In every cases (acidic or
et al. 2002). These methods seem to be inspired basic mobile phases), methods should employ a
from the early works of Renaudin (1984) on the pH which is far from the pKa of the target
separation of ajmalicine, catharanthine, serpen- compounds, since weak variations in the mobile
tine, vindoline and vinblastine. Other studies are phase preparation can change the degree of
suggesting the use of ion-pairing additives such as protonation of the alkaloids, and thus influence
TEA or TFA, which are commonly used for the elution pattern and the reproducibility in the
masking non-derivatized silanol groups (Aerts retention times (Uniyal et al. 2001; McCalley
et al. 1994; St-Pierre and De Luca 1995; Lee and 2002). Uniyal et al. (2001) observed that the
Shuler 2000; Zhao et al. 2001; Uniyal et al. 2001; elution time pattern changed from catharanthine,
El-Sayed et al. 2004). These HPLC methods vincristine, vinblastine and vindoline at pH 6, to
using ion-pairing reagents have been first devel- vincristine, vindoline, catharanthine and vinblas-
oped by Naaranlathi et al. (1987). With a neutral tine at pH 7.5. Using a phosphate buffer at pH 6
pH, ion-exchange interactions and hydrogen with acetonitrile as mobile phase, Tikhomiroff
bonding can occur between dissociated silanol and Jolicoeur (2002) obtained the same elution
and protonated (or partially protonated) alka- sequence than that obtained at pH 7.5 by Uniyal
loids (Nawrocki 1997; McCalley 2002), which et al. (2001). This discrepancy may be explained
usually results in peak tailing. TEA allows com- because Uniyal et al. (2001) used TEA and a
peting alkaloids for the silanol sites. It is usually different column, resulting in a different silanol

123
Phytochem Rev (2007) 6:207–234 227

activity of the column packing. Barthe et al. For both indole and iridoid precursors, an acidic
(2002) found that the elution sequence for vind- aqueous mobile phase (pH 1.8 or 2) was used to
oline and cathranthine was reversed changing the preserve an adequate peak shape for secologanin
mobile phase pH from 5 to 10. Therefore, a (Dagnino et al. 1996). Modulating TCA concen-
judicious combination of buffer and mobile phase tration in Dagnino’s method allowed varying the
pH can be helpful when designing a separation retention time of tryptophan and tryptamine.
method focusing on specific peaks. For instance, Using a similar C18 HPLC column, Tikhomiroff
adjustment of the mobile phase pH at 4.7 instead and Jolicoeur (2002) were able to achieve the
of 3.9 allowed separating strictosidine and non- separation of the indole precursors and secolog-
polar alkaloids from an extract of Tabernaemon- anin in less than 8 min by increasing the flow rate
tana divaricata (Dagnino et al. 1995). Using an without using an ion-pairing molecule. Trypt-
isocratic elution, satisfactory separation of alka- amine can also be analyzed in the same run than
loids can be achieved (Table 7). Bhadra et al. TIAs (Auriola et al. 1989; Moreno et al. 1993;
(1993) and Rijhwani and Shanks (1998) were able Lee-Parsons et al. 2004).
to separate up tofive alkaloids within a unique run
for C. roseus metabolites. Van der Heijden et al. Scaling-up a method
(1987) used an isocratic HPLC method previ-
ously developed for Tabernaemontana species. As a litany stated along this review, the major
Furthermore, separation of the alkaloids can be limitation in developing effective separation and
greatly improved by using a gradient elution identification methods relies in the lack of stan-
(Naaranlahti et al. 1987; Tikhomiroff et al. 2002). dards for most of C. roseus secondary metabo-
It allows achieving a good separation without lites. However, if a lab is conducting metabolic
extending HPLC run time. A typical UV-chro- studies or is screening for therapeutics, it may
matogram obtained from the separation of a become essential to have access to those unavail-
standard mixture of 12 C. roseus secondary able standards. Transfer between academics is a
metabolites using the gradient method developed simple and efficient way but could be difficult
by Hallard et al. (unpublished results reproduced when several milligrams of pure compounds are
with the kind permission of Dr. Verpoorte; see asked. A lab may then be obliged to upscale a
Fig. 4 caption for the complete list of authors), is separation method from the analytical to the
presented in Fig. 4. Some co-eluting peaks cannot semi-preparative (semi-prep) scale. It is thus
be accurately quantified by UV, but they were important to consider this issue already when
discriminated using an ESI-MS detector in single developing an analytical method because some
ion recording mode, based on the mass of the early precautions may save a lot of time and
protonated molecule (not shown). Analysis of a efforts at the further developmental stages,
crude extract of C. roseus hairy roots using the including industrial. The important parameters
gradient method developed by Tikhomiroff and not to forget are the column type and the HPLC
Jolicoeur (2002) is presented in Fig. 5a. All the system supplier. The choice of the column is
alkaloids are eluted within a 23 min run. The important because a rigorous scale-up can be
examples presented in this review show the done more easily using a larger diameter column
advantage of a gradient method for the separation having the same length and packing that the one
of complex mixtures of C. roseus alkaloids which used at analytical scale. Total pressure of the
allow separating up to twelve known compounds system is also an important parameter to consider
in the same run. preventing from packing bed deterioration. Pres-
sure is building-up in the column and the guard-
HPLC methods for the analysis of precursors column and is amplified by the viscosity of the
mobile phase. It is also highly helpful to select a
For the analysis of indole and iridoid precursors flow rate range for the analytical method enabling
(Table 6), Tikhomiroff and Jolicoeur (2002) the use of the same HPLC pump for the semi-
adapted the method of Dagnino et al. (1996). prep method.

123
228 Phytochem Rev (2007) 6:207–234

1000 1000
1 4
a
800 800

3
600 600

mAU

mAU
400 400

2 6 7
5
200 200

0 0

0.0 2.5 5.0 7.5 10.0 12.5 15.0 17.5 20.0 22.5 25.0 27.5 30.0 32.5 35.0
Minutes

1200 1200
1 4
b
1000 1000

3
800 800

mAU
mAU

600 600

400 2 400
5 6 7

200 200

0 0

0.0 2.5 5.0 7.5 10.0 12.5 15.0 17.5 20.0 22.5 25.0 27.5 30.0 32.5 35.0
Minutes

Fig. 5 Analytical method (a) and semi-prep method (b) 20:80; 20–30 min, isocratic elution with a 20:80 mixture; 30–
developed from the modification of the Tikhomiroff and 35 min: isocratic elution 80:20 for column equilibration.1,
Jolicoeur (2002) method. Mobile phase was CH3 COONH4 serpentine; 2, unknown T1; 3, catharanthine; 4, lochneri-
50 mM pH 6 (solvent A) and acetonitrile (solvent B). The cine; 5, unknown T3; 6, tabersonine; 7, unknown T4. The
flow rate was 1 ml min–1 for the analytical method and alkaloids extract was pre-purified using a Strata C18-T SPE
4 ml min–1 for the semi-prep method The eluent profile cartridge (Phenomenex, Torrance, USA) (unpublished
(volume solvent A: volume solvent B) was the same for data)
both methods: 0–20 min, linear gradient from 80:20 to

An example of an analytical method that has semi-preparative run, to preserve similar


been scaled-up to a semi-prep method is given in chromatographic conditions. The run was 7 min
the following. The analytical method developed longer but the separation remained satisfactory
for TIAs by Tikhomiroff and Jolicoeur (2002) was and the pressure limit of the HPLC system was
slightly modified. First, the phosphate buffer was not exceeded. The chromatograms obtained for
changed for ammonium acetate buffer, which is the analytical and semi-prep methods shown to be
more volatile and can thus be easily used in highly similar (Fig. 5). The analytical and semi-
combination to the MS detector for further prep methods used injection volumes of 20 and
characterization of the fractions collected. Sec- 250 ll, respectively. The use of a PDA detector
ond, the flow rate was divided from 2 to 1 ml min– with a larger cell adapted for semi-prep flow
1
. Since the diameter of the semi-prep column was rates explains the similar absorbance in both
2 times that of the analytical one, it was necessary injections. This semi-prep method was designed
to increase the flow rate to 4 ml min–1 for the for the recovery of standards, for both the known

123
Phytochem Rev (2007) 6:207–234 229

molecules as well as for some unidentified alka- of HPLC stationary phase with that resulting
loids. A fraction collector was used and each peak from the application of an electric field (McCalley
was collected based on the slope of the absor- 2002). It has been reported that a C18 stationary
bance signal at 296 nm. The collected fractions phase with a charged surface allows effective
could then be further analyzed for purity through electroosmotic flow and an efficient separation
MS-MS or 1H-NMR detection. Since hundreds (Zimina et al. 1997). However, the limit of
of molecules elute from a single injection, some detection for HPLC analysis is still lower that
compounds may unfortunately co-elute. Thus, that for CE and this is probably due to the low
the use of semi-prep columns may be of inter- sample volume injected in CE (Barthe et al. 2002;
est for pre-fractionation of a complex mixture Dräger 2002).
(Mans et al. 2000). This method allows ‘‘cutting’’
the chromatogram into several fractions contain-
ing, hopefully, one to ten alkaloids each. Future developments
Then, the fractions can be separated under
modified HPLC conditions, and used for bioac- HPLC methods and equipments
tivity screening.
There are many improvements that can be
Other useful analytical techniques achieved to make HPLC an even more powerful
analytical technique. We think that the objective
While TLC was historically seen as an interesting for improving HPLC technique should be to
low cost technique, capillary electrophoresis (CE) simplify method scale-up procedures and enhance
and CZE (capillary zone electrophoresis) is now the capacity for integrating more powerful detec-
gaining in interest. The reader is invited to consult tors. In addition to UV and fluorescence spectra
the review of Stöckigt et al. (2002) on the use of determination, measurement of the molecular
CZE for the analysis of alkaloids. CE allows weight for each peak can be highly useful to
separation of charged molecules based on the analyze plant or cell extracts. Molecular weights
mass-to-charge ratio in an electric field. Due to of C. roseus metabolites have been recently
their high pKa, alkaloids are present in their presented by van der Heijden et al. (2004) in a
protonated form when using an acidic buffer. review inspired from the Dictionary of Natural
CZE was used for the separation of the bisindole Products. The use of MS detection, as TSP-MS
vincristine and vinblastine from a crude leaf (Thermospray), ESI-MS (Electrospray Ionization
extract (Chu et al. 1996). Barthe et al. (2002) Mass Spectrometry) or MALDI-TOF (Matrix
have used a non-aqueous electrophoretic buffer Assisted Laser Desorption with Time-of-Flight)
for separating 11 Vinca alkaloids in less than can enhance the identification of low abundant
10 min. The running buffer consisted in 50 mM compounds and of non-pure peaks with a higher
ammonium acetate and 0.6 M acetic acid in an precision than UV spectra. However, as discussed
organic mixture of methanol–acetonitrile (75:25, previously, because of the silanol activity of
v:v). A constant voltage of 25 kV was applied and HPLC packing, an acidic mobile phase is used
the temperature of the capillary was kept at 20C. to limit peak tailing and ensure batch to batch
Unfortunately, this non-aqueous capillary elec- reproducibility. Although amine modifiers can be
trophoresis method has not been tested on plant included in the mobile phase as masking agents,
extract. their use is not recommended for ESI-MS detec-
Preparative-scale continuous separation of tion since an ion-suppression phenomenon can be
enantiomers by CE has also been reported generated and then greatly reduce the sensitivity
(Spanik 2002) but scale-up potential of CE for of the instrument (Gustavsson et al. 2001; Anti-
phytochemicals has still to be demonstrated gnac et al. 2005). The introduction on the market
(Suntornsuk 2002). Indeed, CE may be an alter- of columns integrating a carbamate internal
native for the analysis of indole alkaloids in crude group in bonded phase seemed to have solved
extracts. This technique combines the selectivity this problem. Peak tailing showed to be reduced

123
230 Phytochem Rev (2007) 6:207–234

for basic compounds as compared to traditional common HPLC methods are based on non-
C18 alkyl packing (O’Gara et al. 1999). The volatile buffers, such as phosphate buffer, which
choice of different MS interfaces for the analysis are not compatible with MS detection (Law and
of alkaloids has been reviewed (Verpoorte and Temesi 2000). Also, since the elucidation of the
Niessen 1994). The use of LC-TSP-MS allowed structure of many alkaloids is achieved by 1H-
quantification of tryptamine, ajmalicine, serpen- NMR, the association of such detection with the
tine, catharanthine and tabersonine (Auriola power of separation of HPLC could be highly
et al. 1989). Hallard (unpublished data, see cap- efficient. Such an approach was presented by
tion of Fig. 4 for complete reference) developed a Bobzin et al. (2000) for the determination of
LC-ESI-MS method allowing quantification of 10 marine alkaloids as inhibitors of an enzyme
alkaloids and 19 precursors. Using a HPLC implicated in the type II diabetes. HPLC-NMR
separation system coupled to an ESI interface, was also used for the screening of plant extracts
Chu et al. (1997) detected vincristine and vin- (Bringmann et al. 1999, 2000, 2002; Zhao et al.
blastine in plant extracts at a limit of detection of 1999). To our knowledge, however, this method
0.2 lg ml–1. The sensitivity for these compounds has never been used with C. roseus crude extracts.
was 2–3 times higher in ESI-MS rather than with Coupling NMR detection to HPLC is though to
UV detection. In addition of increasing the be the unique method for the elucidation of
sensitivity, ESI-MS will help to achieve the isomers.
identification of the numerous compounds which
can not be actually identified from a UV detector
because some secondary metabolites possess Conclusion
highly similar UV-spectra. Zhou et al. (2005)
were able to analyze vindoline, vindolidine, vin- This article has presented a review on the
cristine and vinblastine in a commercial prepara- significant effort of the scientific community to
tion by direct injection ESI-MS-MS. This is develop efficient and reliable HPLC methods for
obviously shortening the time for the identifica- the analysis of C. roseus alkaloids. There are
tion of these molecules. However, the authors some other analytical techniques such as TLC and
mentioned that HPLC separation is required, CE that have also been presented but emphasis
followed by ESI-MS-MS to discriminate catha- has been placed on HPLC because it can be easily
ranthine from the two isomers vindolinine and scaled-up. This article has then reviewed the most
19S-vindolinine, which all have a m/z ratio of 337. recent developments in HPLC analysis of alka-
Maldi-TOF analysis of crude extract represents a loids from C. roseus. The diverse considerations
quick technique for fast screening of different and the crucial steps to achieve an efficient
strains and cell lines for their vinblastine or separation and identification of secondary metab-
vincristine contents, as proposed by Contin and olites were enumerated and discussed. Biomass
van der Heijden (unpublished results, cited in van manipulation, choice of an extraction phase and
der Heijden et al. 2004). However, single quad- of the extraction protocols as well as HPLC
rupole MS detector may not be sufficient for separation and analysis protocols were then
absolute peak identification because many metab- reviewed in details. Furthermore, examples of
olites posses the same molecular weight. For spectra obtained using most common detectors
instance, 13 alkaloids from C. roseus have a were also shown. Finally, suggestions were made
similar molecular weight of 352.432 g mol–1. on how to proceed for developing efficient sep-
Identification of such molecules should be aration and identification methods, based on
achieved after molecular fragmentation in a literature.
tandem mass spectrometer and/or by 1H-NMR The screening of C. roseus secondary metabo-
analysis. Transposition of HPLC methods devel- lites represents a challenge for the pharmaceuti-
oped for UV or fluorescence detector to MS cal industry. The synthesis of the bisindole
detector is possible if the methods are using a alkaloids vinblastine and vincristine may be
volatile buffer as mobile phase. However, most performed industrially through chemical synthesis

123
Phytochem Rev (2007) 6:207–234 231

or from genetically modified plant cells in a near Bhadra R, Shanks JV (1997) Transient studies of nutrient
future. The modification of C. roseus metabolic uptake, growth, and indole alkaloid accumulation in
heterotrophic cultures of hairy roots of Catharanthus
pathways or the insertion of C. roseus genes in roseus. Biotechnol Bioeng 55:527–534
other plant species may also result in an increase Bhadra R, Vani S, Shanks JV (1993) Production of indole
of the biodiversity of the alkaloids obtained (van alkaloids by selected hairy root lines of Catharanthus
der Heijden et al. 2004). Thus, many novel roseus. Biotechnol Bioeng 41:581–592
Blom TJM, Sierra M, Vanvliet TB, Frankevandijk MEI,
unknown alkaloids of therapeutic interest may Dekoning P, Vaniren F, Verpoorte R, Libbenga KR
then be obtained in the future and these new (1991) Uptake and accumulation of ajmalicine into
compounds will have to be separated, identified isolated vacuoles of cultured-cells of Catharanthus
and tested using HPLC. The development of a roseus (L.) G. Don and its conversion into serpentine.
Planta 183:170–177
successful HPLC method is thus a crucial step in Bobzin SC, Yang S, Kasten TP (2000) Application of
the process of identifying the therapeutic liquid chromatography–nuclear magnetic resonance
potential of phytochemicals such as those from spectroscopy to the identification of natural products.
C. roseus. J Chromatogr B 748:259–267
Bringmann G, Messer K, Wohlfarth M, Kraus J, Dumbuya
K, Rückert M (1999) HPLC-CD on-line coupling in
Acknowledgements Authors wish to thank Dr. combination with HPLC–NMR and HPLC–MS/MS
Verpoorte for his kind permission to include in this for the determination of the full absolute stereostruc-
paper his unpublished work shown as Fig. 4. ture of new metabolites in plant extracts. Anal Chem
71:2678–2686
Bringmann G, Wohlfarth M, Heubes M (2000) Observa-
References tion of exchangeable protons by high-performance
liquid chromatography–nuclear magnetic resonance
Abel U, Koch C, Speitling M, Hansske FG (2002) Modern spectroscopy and high-performance liquid chroma-
methods to produce natural-product libraries. Curr tography–electrospray ionization mass spectrometry:
Opin Chem Biol 6:453–458 a useful tool for the hyphenated analysis of natural
Aerts RJ, Gisi D, Decarolis E, Deluca V, Baumann TW products. J Chromatogr A 904:243–249
(1994) Methyl jasmonate vapor increases the devel- Bringmann G, Wohlfarth M, Rischer H, Schlauer J, Reto
opmentally controlled synthesis of alkaloids in Cath- B (2002) Extract screening by HPLC coupled to MS-
aranthus and Cinchona seedlings. Plant J 5:635–643 MS, NMR, and CD: a dimeric and three monomeric
Annesley TM (2003) Ion suppression in mass spectrome- naphthylisoquinoline alkaloids from Ancistrocladus
try. Clin Chem 49:1041–1044 griffithii. Phytochemistry 61:195–204
Antignac JP, Brosseaud A, Gaudin-Hirret I, Andre F, Byun SY, Pedersen H (1994) 2-Phase airlift fermenter
Bizec BL (2005) Analytical strategies for the direct operation with elicitation for the enhanced production
mass spectrometric analysis of steroid and corticoste- of benzophenanthridine alkaloids in cell-suspensions
roid phase II metabolites. Steroids. Web released of Eschscholtzia californica. Biotechnol Bioeng 44:14–
Asada M, Shuler ML (1989) Stimulation of ajmalicine 20
production and excretion from Catharanthus roseus: Capell T, Christou P (2004) Progress in plant metabolic
effects of adsorption in situ, elicitors and alginate engineering. Curr Opin Biotechnol 15:148–154
immobilization. Appl Microbiol Biotechnol 30:475–481 Chen X, Yi C, Yang X, Wang X (2004) Liquid chroma-
Auriola S, Naaranlati T, Lapinjoki SP (1991) Determination tography of active principles in Sophora flavescens
of Catharanthus roseus alkaloids by high-performance root. J Chromatogr B 812:149–163
liquid-chromatography–isotope-dilution thermospray Choi YH, Yoo KP, Kim J (2002) Supercritical fluid
mass spectrometry. J Chromatogr 554:227–231 extraction and liquid chromatography–electrospray
Auriola S, Ranta VP, Naaranlahti T, Lapinjoki SP (1989) mass analysis of vinblastine from Catharanthus roseus.
Thermospray liquid-chromatographic mass-spectro- Chem Pharm Bulletin 50:1294–1296
metric analysis of Catharanthus alkaloids. J Chroma- Chu IH, Bodnar JA, Bowman RN, White EL (1997)
togr 474:181–185 Determination of vincristine and vinblastine in
Balagué C, Wilson G (1982) Growth and alkaloid biosyn- Catharanthus roseus plants by high performance
thesis by cell suspensions of Catharanthus roseus in a liquid chromatography electrospray ionization mass
chemostat under sucrose and phosphate limiting spectrometry. J Chromatogr Relat Technol 20:1159–
conditions. PhysiolVeg 20:515–522 1174
Barthe L, Ribet JP, Pelissou M, Degude MJ, Fahy J, Chu IH, Bodnar JA, White EL, Bowman RN (1996)
Duflos A (2002) Optimization of the separation of Quantification of vincristine and vinblastine in Cath-
vinca alkaloids by nonaqueous capillary electropho- aranthus roseus plants by capillary zone electropho-
resis. J Chromatogr A 968:241–250 resis. J Chromatogr A 755:281–288

123
232 Phytochem Rev (2007) 6:207–234

Collins-Pavao N, Chin CK (1996) Taxol partitioning in Gustavsson SA, Samskog J, Markides KE, Langström B
two-phase plant cell cultures of Taxus brevifolia. J (2001) Studies of signal suppression in liquid chroma-
Biotechnol 49:95–100 tography–electrospray ionization mass spectrometry
Collu G, Bink HHJ, Moreno PRH, Van Der Heijden R, using volatile ion-pairing reagents. J Chromatogr A
Verpoorte R (1999) Determination of the activity of 937:41–47
the cytochrome p450 enzyme geraniol 10-hydroxylase Hendriks MMWB, Cruz-Juarez L, Bont DD, Hall RD
in plants by high-performance liquid chromatography. (2005) Preprocessing and exploratory analysis of
Phytochem Anal 10:314–318 chromatographic profiles of plant extracts. Anal Chim
Contin A, Van Der Heijden R, Lefeber AWM, Verpoorte Acta 545:53–64
R (1998) The iridoid glucoside secologanin is derived Hisiger S, Jolicoeur M (2005) Plant cell culture monitoring
from the novel triose phosphate/pyruvate pathway in using an in situ multiwavelength fluorescence probe.
a Catharanthus roseus cell culture. FEBS Lett Biotechnol Prog 21:580–589
434:413–416 Houghton PJ (2002) Chromatography of the chromone
Dagnino D, Schripsema J, Verpoorte R (1995) Terpenoid and flavonoid alkaloids. J Chromatogr A 967:75–84
indole alkaloid biosynthesis and enzyme-activities in Hughes EH, Hong SB, Gibson SI, Shanks JV, San KY
two cell-lines of Tabernaemontana divaricata. Phyto- (2004) Expression of a feedback-resistant anthranilate
chemistry 39:341–349 synthase in Catharanthus roseus hairy roots provides
Dagnino D, Schripsema J, Verpoorte R (1996) Analysis of evidence for tight regulation of terpenoid indole
several iridoid and indole precursors of terpenoid alkaloid levels. Biotechnol Bioeng 86:718–727
indole alkaloids with a single HPLC run. Planta Med Ingkaninan K, De Best CM, Van Der Heijden R, Hofte
62:278–280 AJP, Karabatak B, Irth H, Tjaden UR, Van Der
De Luca V, Balsevich J, Tyler RT, Eilert U, Panchuk BD, Greef J, Verpoorte R (2000) High-performance liquid
Kurz WGW (1986) Biosynthesis of indole alkaloids: chromatography with on-line coupled UV, mass
developmental regulation of the biosynthetic pathway spectrometric and biochemical detection for identifi-
from tabersonine to vindoline in Catharanthus roseus. cation of acetylcholinesterase inhibitors from natural
J Plant Physiol 125:147–156 products. J Chromatogr A 872:61–73
De Luca V, Fernandez JA, Campbell D, Kurz WGW Jossang A, Fodor P, Bodo B (1998) A new structural class
(1988) Developmental regulation of enzymes of of bisindole alkaloids from the seeds of Catharanthus
indole alkaloid biosynthesis in Catharanthus roseus. roseus: vingramine and methylvingramine. J Org
Plant Physiol 86:447–450 Chem 63:7162–7167
De Luca V, Laflamme P (2001) The expanding universe Klyushnichenko VE, Yakimov SA, Tuzova TP, Syagailo
of alkaloid biosynthesis. Curr Opin Plant Biol 4:225– YV, Kuzovkina IN, Wulfson AN, Miroshnikov AI
233 (1995) Determination of indole alkaloids from R.
De Luca V, St-Pierre B (2000) The cell and developmental serpentina and R. vomitoria by high-performance
biology of alkaloid biosynthesis. Trends Plant Sci liquid-chromatography and high-performance thin-
5:168–173 layer chromatography. J Chromatogr A 704:357–362
Drager B (2002) Analysis of tropane and related alkaloids. Kruczynski A, Hill BT (2001) Vinflunine, the latest Vinca
J Chromatogr A 978:1–35 alkaloid in clinical development: a review of its
Dutta A, Batra J, Pandey-Rai S, Singh D, Kumar S, Sen J preclinical anticancer properties. Oncol/Hematol
(2005) Expression of terpenoid indole alkaloid bio- 40:159–173
synthetic pathway genes corresponds to accumulation Kutney JP, Choi LSL, Kolodziejczyk P, Sleigh SK, Stuart
of related alkaloids in Catharanthus roseus (L) G KL, Worth BR, Kurz WGW, Chatson KB, Constabel
Don. Planta 220:376–383 F (1980) Alkaloid production in Catharanthus roseus
El-Sayed M, Choi YH, Frederich M, Roytrakul S, cell cultures: isolation and characterization of alka-
Verpoorte R (2004) Alkaloid accumulation in Cath- loids from one cell line. Phytochemistry 19:2589–2595
aranthus roseus cell suspension cultures fed with Laflamme P, St-Pierre B, De Luca V (2001) Molecular and
stemmadenine. Biotechnol Lett 26:793–798 biochemical analysis of a madagascar periwinkle root-
Facchini PJ (2001) Alkaloid biosynthesis in plants: bio- specific minovincinine-19-hydroxy-O-acetyltransfer-
chemistry, cell biology, molecular regulation and ase. Plant Physiol 125:189–198
metabolic engineering applications. Annu Rev Plant Law B, Temesi D (2000) Factors to consider in the
Physiol Plant Mol Biol 52:29–66 development of generic bioanalytical high-perfor-
Fahy J, Du Boullay VT, Bigg DCH (2002) New method of mance liquid chromatographic–mass spectrometric
synthesis of vinca alkaloid derivatives. Bioorg Med methods to support drug discovery. J Chromatogr B
Chem Lett 12:505–507 748:21–30
Favali MA, Musetti R, Benvenuti S, Bianchi A, Pressacco Lee CWT, Shuler ML (2000) The effect of inoculum
L (2004) Catharanthus roseus L. plants and explants density and conditioned medium on the production
infected with phytoplasmas: alkaloid production and of ajmalicine and catharanthine from immobilized
structural observations. Protoplasma 223:45–51 Catharanthus roseus cells. Biotechnol Bioeng 67:61–
Gorog S, Herenyi B, Jovanovics K (1977) High-perfor- 71
mance liquid chromatography of Catharanthus alka- Lee-Parsons CWT, Erturk S, Tengtrakool J (2004)
loids. J Chromatogr A 139:203–206 Enhancement of ajmalicine production in Catharan-

123
Phytochem Rev (2007) 6:207–234 233

thus roseus cell cultures with methyl jasmonate is Payne GF, Payne NN, Shuler ML, Asada M (1988) In situ
dependent on timing and dosage of elicitation. Bio- adsorption for enhanced alkaloid production of Cath-
technol Lett 26:1595–1599 aranthus roseus. Biotechnol Lett 10:187–192
Lee-Parsons CWT, Shuler ML (2002) The effect of Pennings EJM, Verpoorte R, Goddijn OJM, Hoge JHC
ajmalicine spiking and resin addition timing on the (1989) Purification of tryptophan decarboxylase from
production of indole alkaloids from Catharanthus a Catharanthus roseus cell-suspension culture. J Chro-
roseus cell cultures. Biotechnol Bioeng 79:408–415 matogr 483:311–318
Mans DRA, Da Rocha AB, Schwartsmann G (2000) Anti- Potier P, Langlois N, Langlois Y, Guéritte F (1975) Partial
cancer drug discovery and development in Brazil: synthesis of vinblastine-like alkaloids. J Chem Soc
targeted plant collection as a rational strategy to 16:670–671
acquire candidate anti-cancer compounds. The Oncol- Rao SR, Ravishankar GA (2002) Plant cell cultures:
ogist 5:185–198 chemical factories of secondary metabolites. Biotech-
McCalley DV (2002) Analysis of the Cinchona alkaloids by nol Adv 20:101–153
high-performance liquid chromatography and other Renaudin JP (1985) Extraction and fluorimetric detection
separation techniques. J Chromatogr A 967:1–19 after high-performance liquid chromatography of
Meijer AH, Verpoorte R, Hoge JHC (1993) Regulation of indole alkaloids from cultured cells of Catharanthus
enzymes and genes involved in terpenoid indole roseus. Physiol Veg 23:381–388
alkaloid biosynthesis in Catharanthus roseus. J Plant Renaudin JP (1984) Reversed-phase high-performance
Res 3:145–164 liquid chromatographic characteristics of indole alka-
Molyneux RJ, Gardner DR, James LF, Colegate SM loids from cell suspension cultures of Catharanthus
(2002) Polyhydroxy alkaloids: chromatographic anal- roseus. J Chromatogr A 291:165–174
ysis. J Chromatogr A 967:57–74 Renaudin JP (1989) Different mechanisms control the
Monforte-Gonzáles M, Ayaora-Talavera T, Maldonado- vacuolar compartmentation of ajmalicine in Catha-
Mendoza IE, Loyola-Vargas VM (1992) Quantitative ranthus roseus cell cultures. Plant Physiol Biochem
analysis of serpentine and ajmalicine in plant tissues 27:613–621
of Catharanthus roseus and hyoscyamine and scopol- Renaudin JP, Brown SC, Guern J (1982) Compartmenta-
amine in root tissues of Datura stramonium by thin tion mechanisms of indole alkaloids in cell suspension
layer chromatography–densitometry. Phytochem of Catharanthus roseus. Phyisol Veg 20:533–547
Anal 3:117–121 Rijhwani SK, Shanks JV (1998) Effect of elicitor dosage
Moreno PRH, Van der Heijden R, Verpoorte R (1993) and exposure time on biosynthesis of indole alkaloids
Effect of terpenoid precursor feeding and elicitation by Catharanthus roseus hairy root cultures. Biotechnol
on formation of indole alkaloids in cell-suspension Prog 14:442–449
cultures of Catharanthus roseus. Plant Cell Rep Rodriguez S, Compagnon V, Crouch NP, St-Pierre B, De
12:702–705 Luca V (2003) Jasmonate-induced epoxidation of
Moreno-Valenzuela OA, Galaz-Avalos RM, Minero-Garcia tabersonine by a cytochrome p-450 in hairy root
Y, Loyola-Vargas VM (1998) Effect of differentiation on cultures of Catharanthus roseus. Phytochemistry
the regulation of indole alkaloid production in Catha- 64:401–409
ranthus roseus hairy roots. Plant Cell Rep 18:99–104 Satdive RK, Fulzele DP, Eapen S (2003) Studies on
Morgan JA, Barney CS, Penn AH, Shanks JV (2000) production of ajmalicine in shake flasks by multiple
Effects of buffered media upon growth and alkaloid shoot cultures of Catharanthus roseus. Biotechnol
production of Catharanthus roseus hairy roots. Appl Prog 19:1071–1075
Microbiol Biotechnol 53:262–265 Schroder G, Unterbusch E, Kaltenbach M, Schmidt J,
Morgan JA, Shanks JV (2002) Quantification of metabolic Strack D, De Luca V, Schroder J (1999) Light-
flux in plant secondary metabolism by a biogenetic induced cytochrome P450-dependent enzyme in in-
organizational approach. Metabol Eng 4:257–262 dole alkaloid biosynthesis: tabersonine 16-hydroxy-
Naaranlahti T, Nordstrom M, Huhtikangas A, Lounasmaa lase. FEBS Lett 458:97–102
M (1987) Determination of Catharanthus alkaloids by Shanks JV, Bhadra R, Morgan J, Rijhwani S, Vani S
reversed-phase high-performance liquid chromatog- (1998) Quantification of metabolites in the indole
raphy. J Chromatogr A 410:488–493 alkaloid pathways of Catharanthus roseus: implica-
Naaranlahti T, Ranta V-P, Jarho P, Nordström M, tions for metabolic engineering. Biotechnol Bioeng
Lapinjoki SP (1989) Electrochemical detection of 58:333–338
indole alkaloids of Catharanthus roseus in high- Sim SJ, Chang HN, Liu JR, Jung KH (1994) Production
performance liquid chromatography. Analyst and secretion of indole alkaloids in hairy root cultures
114:1229–1331 of Catharanthus roseus – effects of in-situ adsorption,
Nawrocki J (1997) The silanol group and its role in liquid fungal elicitation and permeabilization. J Ferment
chromatography. J Chromatogr A 779:29–71 Bioeng 78:229–234
O’Gara JE, Walsh DP, Alden BA, Casellini P, Walter TH Singh D, Maithy A, Verma R, Gupta M, Kumar S (2000)
(1999) Systematic study of chromatographic behavior Simultaneous determination of Catharanthus alka-
vs. alkyl chain length for HPLC bonded phases loids using reversed phase high performance liquid
containing an embedded carbamate group. Anal chromatography. J Liquid Chromatogr Relat Technol
Chem 71:2992–2997 23:601–607

123
234 Phytochem Rev (2007) 6:207–234

Spanik I, Lim P, Vigh G (2002) Use of full-column Van Der Heijden R, Jacobs DI, Snoeijer W, Hallared D,
imaging capillary isoelectric focusing for the rapid Verpoorte R (2004) The Catharanthus alkaloids:
determination of the operating conditions in the pharmacognosy and biotechnology. Curr Med Chem
preparative-scale continuous free-flow isoelectric 11:607–628
focusing separation of enantiomers. J Chromatogr Van Der Heijden R, Lamping PJ, Out PP, Wijnsma R,
A 960:241–246 Verpoorte R (1987) High-performance liquid chro-
St-Pierre B, De Luca V (1995) A cytochrome p-450 matographic determination of indole alkaloids in a
monooxygenase catalyzes the first step in the conver- suspension culture of Tabersnaemontana divaricata. J
sion of tabersonine to vindoline in Catharanthus Chromatogr 396:287–295
roseus. Plant Physiol 109:131–139 Van Elswijk DA, Irth I (2003) Analytical tools for the
St-Pierre B, Vazquez-Flota FA, De Luca V (1999) detection and characterization of biologically active
Multicellular compartmentation of Catharanthus ro- compounds from nature. Phytochem Rev 1:427–439
seus alkaloid biosynthesis predicts intercellular trans- Vazquez-Flota F, Carrillo-Pech M, Minero-Garcia Y, de
location of a pathway intermediate. Plant Cell 11:887– Lourdes Miranda-Ham M (2004) Alkaloid metabo-
900 lism in wounded Catharanthus roseus seedlings. Plant
Stevens LH, Giroud C, Pennings EJM, Verpoorte R Physiol Biochem 42:623–628
(1993) Purification and characterization of strictosi- Verpoorte R, Memelink J (2002) Engineering secondary
dine synthase from a suspension culture of Cinchona metabolite production in plants. Curr Opin Biotech
robusta. Phytochemistry 33:99–106 13:181–187
Stöckigt J, Sheludko Y, Unger M, Gerasimenko I, War- Verpoorte R, Niessen W (1994) Liquid chromatography
zecha H, Stöckigt D (2002) High-performance liquid coupled with mass spectrometry in the analysis of
chromatographic, capillary electrophoretic and capil- alkaloids. Phyotchem Anal 5:217–232
lary electrophoretic–electrospray ionisation mass Verpoorte R, Van Der Heijden R, Ten Hoopen HJG,
spectrometric analysis of selected alkaloid groups. J Memelink J (1999) Metabolic engineering of plant
Chromatogr A 967:85–113 secondary metabolite pathways for the production of
Strege M (1999) High-performance liquid chromato- fine chemicals. Biotechnol Lett 21:467–479
graphic–electrospray ionization mass spectrometric Whitmer S, Van Der Heijden R, Verpoorte R (2002)
analyses for the integration of natural products with Effect of precursor feeding on alkaloid accumulation
modern high-throughput screening. J Chromatogr B by a strictosidine synthase over-expressing transgenic
725:67–78 cell line s1 of Catharanthus roseus. Plant Cell Tiss Org
Suntornsuk L (2002) Capillary electrophoresis of phyto- Cult 69:85–93
chemical substances. J Pharm Biomed Anal 27:679– Wong PL, Royce AJ, Lee-Parsons CWT (2004) Improved
698 ajmalicine production and recovery from Catharan-
Tikhomiroff C, Allais S, Klvana M, Hisiger S, Jolicoeur M thus roseus suspensions with increased product re-
(2002) Continuous selective extraction of secondary moval rates. Biochem Eng J 21:253–258
metabolites from Catharanthus roseus hairy roots with Zhao Y, Nookandeh A, Schneider B, Sun X, Schmitt B,
silicon oil in a two-liquid-phase bioreactor. Biotechnol Stöckigt J (1999) Lignans from Torreya jackiiidenti-
Progr 18:1003–1009 fied by stopped-flow high-performance liquid chro-
Tikhomiroff C, Jolicoeur M (2002) Screening of Catha- matography–nuclear magnetic resonance
ranthus roseus secondary metabolites by high-perfor- spectroscopy. J Chromatogr A 837:83–91
mance liquid chromatography. J Chromatogr A Zhao J, Zhu WH, Hu Q (2001) Enhanced catharanthine
955:87–93 production in catharanthus roseus cell cultures by
Toivonen L, Ojala M, Kauppinen V (1991) Studies on the combined elicitor treatment in shake flasks and
optimization of growth and indole alkaloid production bioreactors. Enzyme Microbiol Technol 28:673–681
by hairy root cultures of Catharanthus roseus. Bio- Zhou H, Tai YP, Sun CR, Pan YJ (2005) Rapid identifica-
technol Bioeng 37:673–680 tion of Vinca alkaloids by direct-injection electrospray
Uniyal GC, Bala S, Mathur AK, Kulkarni RN (2001) ionisation tandem mass spectrometry and confirmation
Symmetry C18 column: a better choice for the analysis by high-performance liquid chromatography–mass
of indole alkaloids of Catharanthus roseus. Phytochem spectrometry. Phytochem Anal 16:328–333
Anal 12:206–210 Zimina TM, Smith RM, Myers P (1997) Comparison of
Van Der Heijden R, Verpoorte R, Ten Hoopen JG (1989) ODS-modified silica gels as stationary phases for
Cell and tissue cultures of Catharanthus (L.)G. Don: a electrochromatography in packed capillaries. J Chro-
literature survey. Plant Cell Tiss Org 18:231–280 matogr A 758:191–197

123

View publication stats

You might also like