Download as pdf or txt
Download as pdf or txt
You are on page 1of 44

Journal of Ethnopharmacology 284 (2022) 114647

Contents lists available at ScienceDirect

Journal of Ethnopharmacology
journal homepage: www.elsevier.com/locate/jethpharm

Catharanthus roseus (L.) G. Don: A review of its ethnobotany,


phytochemistry, ethnopharmacology and toxicities
Sunil Kumar a, Bikarma Singh b, c, *, Ramesh Singh d
a
Department of Chemistry, Ma. Kanshiram Government Degree College, Ninowa, (affiliated to Chhatrapati Shahu Ji Maharaj University (CSJM) Kanpur), Farrukhabad,
209602, Uttar Pradesh, India
b
Botanic Garden Division, CSIR-National Botanical Research Institute, Rana Pratap Marg, Lucknow, 226001, Uttar Pradesh, India
c
Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
d
Department of Botany, Government Degree College Bahua Dehat, (affiliated to Professor Rajendra Singh (Rajju Bhaiya) University Prayagraj), Fatehpur, 212663, Uttar
Pradesh, India

A R T I C L E I N F O A B S T R A C T

Keywords: Ethnopharmacological relevance: Catharanthus roseus (L.) G. Don is a well known medicinal plant belonging to
Madagascar periwinkle Or periwinkle plant family Apocynaceae that have been traditionally used as medicine since ancient times. C. roseus is a well-
Vinca alkaloids recognized herbal medicine due to its anticancer bisindole alkaloids (vinblastine (111), vincristine (112) and
Traditional medicine
vindesine (121)). In the Ayurvedic system of medicine, different parts of C. roseus are used in folklore herbal
Anticancer activity and quality control
medicine for treatment of many types of cancer, diabetes, stomach disorders, kidney, liver and cardiovascular
Monoterpene indole alkaloids /bisindole
alkaloids diseases.
Analytical methods. pharmacological activity Aim of the study: The main idea behind this communication is to update comprehensively and analyze critically
Toxicology the traditional applications, phytochemistry, pharmacological activities, and toxicity of various extracts and
isolated compounds from C. roseus.
Materials and methods: The presented data covers scientific works on C. roseus published across the world between
1967 and 2021 was searched from various international publishing houses using search engines as well as several
traditional texts like Ayurveda and relevant books. Collected data from different sources was comprehensively
summarized/analyzed for ethnomedicinal uses, phytochemistry, analytical chemistry, biological activities and
toxicity studies of C. roseus.
Results and discussion: C. roseus has a wide range of applications in the traditional system of medicine especially in
cancer and diabetes. During phytochemical investigation, total of 344 compounds including monoterpene indole
alkaloids (MIAs) (110), bisindole alkaloids (35), flavonoids (34), phenolic acids (9) and volatile constituents
(156) have been reported in the various extracts and fractions of different plant parts of C. roseus. The extracts
and isolated compounds of C. roseus have to exhibit many pharmacological activities such as anticancer/cyto­
toxic, antidiabetic, antimicrobial, antioxidant, larvicidal and pupicidal. The comparative toxicity of extracts and
bioactive compounds investigated in dose dependent manner. The investigation of toxicity showed that the both
extracts and isolated compounds are safe to a certain limit beyond that they cause adverse effects.
Conclusion: This review is a comprehensive, critically analyzed summarization of sufficient baseline information
of selected topics in one place undertaken till date on C. roseus for future works and drug discovery. The
phytochemical investigation including biosynthetic pathways showed that the MIAs and bisindole alkaloids are
major and characteristic class of compounds in this plant. The present data confirm that the extracts/fractions
and their isolated alkaloids especially vinblastine (111) and vincristine (112) have a potent anticancer/cytotoxic
and antidiabetic property and there is a need for further study with particular attention to the mechanisms of
anticancer activity. In biosynthesis pathways of alkaloids especially bisindole alkaloids, some enzymes and
rearrangement are unexposed therefore it is required to draw special attention. It also focuses on attracting the
attention of scientific communities about the widespread biological activities of this species for its better utili­
zation prospects in the near future.

* Corresponding author. Botanic Garden Division, CSIR-National Botanical Research Institute, Rana Pratap Marg, Lucknow, 226001, Uttar Pradesh, India.
E-mail address: drbikarma.singh@nbri.res.in (B. Singh).

https://doi.org/10.1016/j.jep.2021.114647
Received 30 April 2021; Received in revised form 7 September 2021; Accepted 14 September 2021
Available online 22 September 2021
0378-8741/© 2021 Elsevier B.V. All rights reserved.
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

1. Introduction on C. roseus are identified while preparing the comprehensive data and
presented for the speed-up drug discovery process. Furthermore, this
Plant as a natural medicine has been used by the people since ancient review also focuses on attracting the attention of scientific communities
times (Singh, 2020). Catharanthus roseus (L.) G.Don belongs to the about the widespread biological activities of this species of plant for its
dogbane family Apocynaceae is one of the most explored short-lived better utilization prospects in the near future. Therefore, the present
medicinal plant species popularly called “Sadabahar” in some Asian review aims to provide comprehensive, critically analyzed and authentic
countries such as India, Pakistan and Nepal (Jacobs et al., 2004; Naeem data on botanical description, traditional medicinal applications,
et al., 2019). This plant species is native to Madagascar. C. roseus is phytochemistry, biosynthesis, separation and analytical techniques,
usually grown as medicinal as well as ornamental plants in parks, gar­ pharmacological properties of extracts/active constituents and toxicity
dens, farms and to beautify landscape places (Adekomi, 2020). C. roseus of extracts/bioactive compounds of C. roseus till date.
has an ancient past in Asian and African countries where this plant An extensive survey of literature related to C. roseus was conducted
species was planted near temples and gardens (Shukla et al., 2010). The up to August 2021. The relevant literary works published on this plant
different plant parts of C. roseus are used traditionally in the Indian species related to botany, ethnomedicinal or traditional uses, phyto­
system of medicine such as Ayurveda, Siddha, Unani and traditional chemical constituents, pharmacological activities, and clinical studies
Chinese medicine (TCM) (Das and Sharangi, 2017). It is employed for were collected from traditional Chinese medicine (TCM), Ayurveda, and
the treatment of cancer, genitourinary system, kidney, liver, cardio­ research articles across the world using a combination of the following
vascular diseases (Zhu et al., 2015). The flower is used due to their keywords: “C. roseus”, “bright eyes”, “cape periwinkle”, “graveyard
fragrance and brain-stimulating odour. The extracts of different parts plant”, “Madagascar periwinkle”,“old maid”, “pink periwinkle”, “rose
and its fractions, and isolated characterized compounds found to have periwinkle”, “medicinal uses of C. roseus”, “phytochemicals of
various pharmacological properties such as antimicrobial, antioxidant, C. roseus”, “pharmacological activities of C. roseus”, “morphology,
anthelmintic, antifeedant, antisterility, antidiarrheal and antidiabetic description, taxonomy or botany of C. roseus”, synonyms as “Ammocallis
(Gajalakshmi et al., 2013). rosea”, “Lochnera rosea”, ‘Pervinca rosea”, ‘Vinca rosea”, “V. speciosa”,
In the last four decades, researchers across the globe showed much various other accepted infra-specifics and synonyms of the plant were
interest in the C. roseus because it contains anticancer bisindole/ also used but the maximum results were obtained from the above key­
monoterpene indole alkaloids (MIAs) and several of these known com­ words. The electronic databases used for the collection of relevant in­
pounds exhibits very strong and potent pharmacological properties formation of C. roseus include Google Scholar, Web of Science, Scopus,
(Dutta et al., 2005; Mishra and Verma, 2017; Barkat et al., 2017; Kumar PubMed, Science Direct, Taylor and Francis online, Springer Link, Na­
et al., 2018a). The marker compounds vinblastine (111), vincristine tional Center for Biotechnology Information (NCBI), Chemspider, and
(112) vindesine (121)and vinorelbine (143) are characteristic bisindole SciFinder. The mentioned databases were used to collect detailed in­
alkaloids that have been permitted for clinical use in the U.S. Food and formation about the common names, botanical characteristics,
Drug Administration for chemotherapy and other medicinal use in the geographical distribution, ethnobotany, phytochemistry, pharmacolog­
pharma sectors (Tsuruo et al., 1981; Osterlind et al., 1982; Pennanen ical aspects, clinical studies, and toxicity on C. roseus. All presented
and Huhtikangas, 1990; Kumar et al., 2018b; Al-Quteimat, 2020). These structures of C. roseus natural products/compounds were drawn using
compounds (111, 112 and 121) are more expensive and have high de­ ChemDraw Pro 8.0 software (PerkinElmer). The PubChem database was
mand in the global market. Besides, there are several other limited used to check updated IUPAC names of phytochemicals reported for the
shoot-specific bioactive compounds present in this plant species which plant.
are found to have medicinal use in pharmaceutical sectors (Yamamoto
et al., 2016). The market prices for compounds 111 and 112 are esti­ 2. Botanical aspects
mated at $2 million/kg, and $15 million/kg, respectively. The supply of
both compounds 111 and 112 are limited by the availability of the plant 2.1. Taxonomy and classification
worldwide. The annual market value of compound 111 and 112 are
about $1,000,000–3,500,000 per kg. The costs of velbane and velsar are C. roseus plant species belongs to the order of ranking of Gentianales.
made from vinblastine sulfate $ ̃ 2/mg or $ 2 million/kg and are used in The member species of Apocynaceae family recorded to occur in trop­
the treatment of many types of cancer like testicular cancer, Hodgkin’s ical, subtropical, and temperate biogeographic zones, mostly consisting
disease and Kaposi’s sarcoma. Similarly, vincristine sulfate based drugs of deciduous trees, shrubs and herbs (Bihani, 2021). C. roseus is taxo­
(Oncovin and Vincasar PFS) costs ̃$ 15/mg or $ 15 million/kg and are nomically placed in the Kingdom Plantae, Class Magnoliopsida, Order
used for the treatment of leukemia in children (Alam and Sharaf-Eldin, Gentianales, Family Apocynaceae and Genus Catharanthus (Integrated
2016). Taxonomic Information System). There are about 359 genera in the
The significance of C. roseus in the modern system of medicine has family and 9 (accepted) species for the genus Catharanthus (Plants of the
been recognized after innovative characterization of anticancer bisin­ Whole Online). The various vernacular names of C. roseus are given in
dole alkaloids such as compounds 111 and 112, which mainly present in Table 1. The synonyms of C. roseus include: Ammocallis rosea (L.) Small,
its leaves. The isolation and purification process of both compounds 111 Lochnera rosea (L.) Rchb. ex Spach, Pervinca rosea (L.) Gaterau, Vinca
and 112 from leaves are very tedious, time-consuming and costly due to rosea L. and Vinca speciosa Salisb.
the presence of very low content of these compounds. Recently, many
advanced and high-throughput separation analytical techniques have 2.2. Morphology
been developed for isolation, purification, identification, characteriza­
tion and quantitation of crude extract prepared from C. roseus (Lin et al., C. roseus recorded to have various botanical characteristics that
2015; Parthasarathy et al., 2020). differentiate this species from other plants within the genera and family.
Most of the review articles on C. roseus have been published in 2007 The plant grows as a perennial herb or undershrub, up to 60 cm tall,
on biosynthesis of MIAs and pharmacological properties (Hisiger and woody base, and milky latex sap (Allorge et al., 2015). The leaves are
Jolicoeur, 2007; Memelink and Gantet, 2007; MustafaVerpoorte, 2007; petiolated, shape oblong, and texture leathery, whereas the arrangement
Oudin et al., 2007; Pietrosiuk et al., 2007: Piovan and Filippini, 2007; of leaves are opposite as well as alternate (Bertaccini, 2007; Junaid
Zárate, and Verpoorte, 2007; Zhao and Verpoorte, 2007; Gajalakshmi et al., 2007). The flowers usually arise as single (Mishra et al., 2001),
et al., 2013; Zhu et al., 2015; Nisar et al., 2016; Pan et al., 2016: Das and attractive in colours which can be pink, white, or a mixture of both,
Sharangi, 2017; Mahroug et al., 2007; Mishra and Verma, 2017; Sen­ sometimes the pale pink with a dark violet dot in the center with many
bagalakshmi et al., 2017; Paarakh et al., 2019). The gaps of knowledge hues and having spreading five petals (Singh et al., 2008). Based on

2
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Table 1 flowers, roots) of this species have been employed for the treatment of
Vernacular names of C. roseus across the globe. diseases in various countries (Table 2). In Ayurveda, stem, leaves, and
S. Country Common name Reference roots of this plant species are employed to cure athelmintic, emetic,
N. digestive, hypotensive, laxative, sedative, stomachic, toothache, dia­
Bangladesh Nayantara (ক্যাথারান্থস) Islam et al. (2009a) betes, asthma, gastrointestinal ailments, and problems in female sexu­
Brazil Boa-noite, Congorca Arora et al. (2010) ality disorders (Rahman et al., 2004; Aslam et al., 2010; Gajalakshmi
Cook Islands Tiare-tupapaku-kimo (Arora et al. (2010) et al., 2013; Sagar et al., 2020). Few studies such as Nammi et al. (2003)
Dominica Caca poule Senbagalakshmi et al. and Al-Quteimat (2020) reported that eighteen leaves and some roots of
(2017)
French Pervenchede de Madagascar Hiruki & da Rocha
the plant boiled in a kettle of water taken in the Cook Islands and in West
Guiana (1986) Indies to control diabetes in the form of infused whiskey. C. roseus is also
Guatemala Chatilla Arora et al. (2010) utilised in folklore traditional medicine system to control menorrhagia,
Guyana, Periwinkle Marcone et al. (1997); toothache, high blood pressure, memory loss, and diarrhoea in various
Jamaica, USA Arora et al. (2010)
regions of India (Srinivasan et al., 2001; Govindasamy and Srinivasan,
India Ainskati, Billaganneru, Retna and Ethalsa
Nayantara, Nityakalyani, (2013) 2012; Rasineni et al., 2010). In India, mainly in the state of Odisha and
Periwinkle, Sadabahar, Arora et al. (2010) Assam, the fresh juice extracted from C. roseus leaves used to treat sting
Sadaphul, Ushamanjairi bites of wasp, whereas the root parts employed in the treatment of
Japan Nichinich-so, Nichinichi-so Arora et al. (2010) cancer. Besides, the C. roseus leaves is used to cure menorrhagia, bone
Kenya Maua Senbagalakshmi et al.
(2017); Arora et al.
disorders, hypertension, skin infections, diarrhoea/dysentery, dengue
(2010) fever, malaria, toothache, eye irritation, and respiratory problems in
Madagascar Madagascar periwinkle, Bright Arora et al. (2010) Thailand, Brazil, USA, Australia and China (Mehta et al., 2013; Kabesh
eye, Old maid, Pink perwinkle et al., 2015; Vipasha et al., 2016; Renjini et al., 2017). The European
Mexico Ninfa Arora et al. (2010)
people use this plant species for minor illnesses such as headaches and
Pakistan Sada-bahar Mishra and Verma
(2017) manage diabetes naturally. The roots have been used for the manage­
Peru Chavelita Arora et al. (2010) ment of hypertension due to their sedative and tranquilizing properties;
Philippines Atay-biya, Chichirica, Senbagalakshmi et al. similar compounds present in C. roseus also reported from different
Kantotan, Periwinkle, (2017); Arora et al. species of Rauwolfia as the tonic for general debility (Kumar et al. 2016a,
Tsitsirika (2010)
2016b). In Madagascar, leaves of C. roseus are employed as an emetic
Rodrigues Saponaire Gurib-Fakim et al.
Islands (1996) drug. Similarly, the underground root plant parts have been used as
Sri Lanka Minimal, Patti-poo Senbagalakshmi et al. laxative, and for curing toothache (Nejat et al., 2015). Young leaves
(2017); Arora et al. employed for the treatment of cramps in stomach and the decoction
(2010)
prepared from roots are used to removes the intestinal parasites in
Thailand Phaeng phoi farang, Phang- Arora et al. (2010)
puai- fa-rang Philippines. The leaves infusion employed for indigestion and dyspepsia
Venda Liluvha Arora et al. (2010) in Mauritius. In many countries, C. roseus floral extracts have been used
Vietnam Dua can for the treatment of dysmenorrhea (Indo-China), an eye-wash for infants
West Indies Brown man’s fancy, Arora et al. (2010) (Cuba and Jamaica), asthma/ respiratory problem (Bahamas), hyper­
Consumption bush, Old maid,
tension (Bermuda), malaria, menstrual problems and diaphoresis (Sur­
Periwinkle, Pink flower, Ram
goat rose, Red rose, Sailor’s inam and Caribbean) (Aslam et al., 2010; Senbagalakshmi et al., 2017).
flower, White tulip The extracts or infusions of leaves of C. roseus is used by the people of
Uganda to cure ulcers associated with digestive tracts, whereas the dried
leaf powder with milk helpful for the treatment of full-grown abscesses
variation in flower colour, there are two varieties of C. roseus. The one in Botswana (Naeem et al., 2019). The root decoction is also used to cure
producing pink flower is called the “Rosea” and the other having the dysmenorrhea in Togo (Senbagalakshmi et al., 2017). In China, this
white flowers is known as “Alba” (Sagar et al., 2020; Al-Quteimat, 2020) species is widely used to cure blood cancer, cough and Brill Symmers
(Fig. 1). Fruits comprised of two narrow cylindrical follicles having (Senbagalakshmi et al., 2017). Correspondingly, Central and South
numerous seeds. America’s people accepted this herb as the plant species for lungs
congestion and throat infections (Don, 1999; Ross, 2003; Das and
2.3. Geographic distribution Sharangi, 2017; Mishra and Verma, 2017; Renjini et al., 2017; Senba­
galakshmi et al., 2017). The tea prepared from leaves of the plant has
The natural wild habit of C. roseus is Madagascar and in the due been used to treat the both mouth and gastric ulcers in Asian countries
passage of time, the plant got distributed across the globe either as an (Rambhai et al., 2019; Tabuti et al., 2003).
ornamental plant or as introduced species for medicinal purposes (Gupta
et al., 2007: Aruna et al., 2015; Falcão et al., 2017). The wide range of 4. Phytochemistry
distribution in various countries recorded for this species is presented in
Table 1. Tong et al. (2011) reported C. roseus as an evergreen herbaceous 4.1. Biosynthesis and synthetic biology advances
ornamental plant of tropical countries. Total nine species reported under
the genus, out of which 7 species known from Madagascar; one from Synthetic biology play an important role in biosynthesis of MIAs and
India and Sri Lanka, while one species is cultivated in China (El-Ghazaly, bisindole alkaloids via combination of various disciplines including
1990; Allorge et al., 2015). C. roseus is more commonly known as molecular biology, molecular engineering, mathematics and physics to
Madagascar periwinkle. It has a side effects on skin such as warm design and build novel proteins, genetic circuits and metabolic networks
prickling and hallucinations (Das and Sharangi, 2017). (McDaniel and Weiss, 2005). Biosynthesis of assembly of these MIAs and
bisindole alkaloids is a multi-step and enzyme-catalyzed process where
3. Traditional uses and ethnopharmacology more than one substrate is converted into more complex metabolites in
parts of C. roseus (Fig. 2). The leaf epidermis is enriched with synthetic
C. roseus has been of prime importance in the traditional medicine enzymes where most of the MIAs and bisindole alkaloids biosynthesis
systems and has wide therapeutic applications for many centuries due to are expressed. 3α (S)-strictosidine (105) is an important intermediate in
its unpleasant side effects. The different plant parts (leaves, stems, biosynthesis of most of MIAs, which is a condensation product of

3
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Fig. 1. Morphology of C. roseus, (A) habit of the whole plant, (B) close view of stem and leaves, (C) root parts, (D–E) close view of flowers, (F) fruits, (G) seeds.

secologanin and tryptamine. Vindoline (27) and catharanthine (55), 2019). The compounds and their corresponding classes are presented in
which are produced by compound 105 via multi-step enzymatic reac­ Table 3.
tion, can form α-3, 4-anhydrovinblastine by the condensation reaction.
In biosynthesis of bisindole alkaloids like compounds 111 and 112 4.2. Monoterpene indole alkaloids (MIAs)
formed by togather combination of compounds 55 and 27 are modified
and converted into iminium ions. Both iminium ions and 3′ ,4′ -anhy­ Total of 106 MIAs (1–106) and three indole derivatives (107–109)
drovinblastine (117) are interconvertible to each other and final and one piperidine derivatives (110) have been reported from different
biosynthetic product is compound 111. Furthermore, compounds 112 parts (leaves, roots, stems) of C. roseus and tissue culture of leaves, roots,
and leurosine (125) are synthesized from compound 111 but convertible and stems (Wang et al., 2012a). Biosynthetic pathways showed that the
enzymes involved during the biosynthesis are unknown. Only 26 genes combination of tryptophan and serotonin produced compound 105 and
involved in the assembly of these two bisindole alkaloids are known, two further many enzymatic reactions synthesized an intermediate or pre­
key reactions have eluded characterization to complete the documen­ cursor either vincoside (96) or compound 105 or both simultaneously.
tation of the compound 111 pathway in C. roseus (Memelink and Gantet, Compounds 96 and 105 synthesized a corynantheine class of interme­
2007; Zhu et al., 2015; Mahroug et al., 2007; Quan et al., 2019). diate (97–104 and 106) due to cyclization of tepene ring in different
Many transcription factors are involved in the regulation of MIAs manner which were detected and isolated from leaves (O’Connor and
biosynthesis, such as apetala2/ethylene responsive factor (AP2/ERF) Maresh, 2006; O’Connor et al., 2012). Corynantheine is an intermediate
and WRKY. The illustration of biosynthetic pathways have laid a basis for synthesis of sarpagine (61–71), ajmaline (72–75), reserpine (76–79)
for the study of synthetic biology. Recently, compounds 27 and 105 and ajmalicine (80–95) classes of MIAs. The cyclization and rear­
have been synthesized in heterologous hosts saccharomyces cerevisiae. rangement of corynantheine generated three classes of MIAs namely
Research about synthetic biology and the regulation mechanisms will vincadifformine (1–26), tabersonine (27–51) and catharanthine (52 and
provide guidance for the production and development of MIAs and 53). Oxidation and reduction at C-2 and C-3 by both directions i.e. front
bisindole alkaloids drugs in C. roseus (Oudin et al., 2007; Zárate, and and back with the help of enzymes produced many MIAs of classes
Verpoorte, 2007; Pan et al., 2016). vincadifformine (1) and tabersonine (6). Similar circumstance occurred
Total of 345 compounds including alkaloids, phenolics and volatile with the C-6 and C-7 tabersonine (6–26) and vindoline (27–51) with
compounds have been categorized and summarized in the review. The enzymatic reduction (SN 15) oxidation (16–20) and epoxidation
bisindole alkaloids (compounds 111), 112, 121), vincovalinine (137), (16–20) of double bond of terpene. The cyclization of terpenine moiety
vincovalicine (138), vincadioline (139)) and MIAs (compounds 27, 55, is synthesizes a variety of complex skeletons 46–52 and 54–60). Most of
vinpocetine (52), trichophylline (54), vincarodine (56), and pleiocarp­ the sarpagine (65), ajmaline (72), ajmalicine (80) and reserpine (78) of
amine (57)), are characteristic as a major/characteristic compounds MIAs have been reported from C. roseus and Rauwolfia species (Kumar
which were isolated, identified, and chemically characterized from et al., 2016a, 2016b, Baig et al., 2021) (Fig. 3Aand 3B).
different parts especially leaves of C. roseus (Rao and Ahmed, 2014;
Lawal et al., 2015; Nisar, 2017a, 2017b; Kumar et al., 2018a). Most of
4.3. Bisindole alkaloids
flavonoids, phenolic acids, triterpenes and sterols have been identified
and characterized by liquid chromatography–mass spectrometry
A total of 35 (111–145) bisindole alkaloids have been reported in
(HPLC-MS) and liquid chromatography with tandem mass spectrometry
different parts of C. roseus and its suspension cell cultures (Wang et al.,
(HPLC-MS/MS), whereas gas chromatography–mass spectrometry
2012b, 2014) (Fig. 4A and4B). Biosynthetically, the bisindole alkaloids
(GC-MS) are used for the analysis of volatile chemical constituents from
(like (111) and 112) synthesized via combination of two different and
extracts of C. roseus parts (Jeong and Lim, 2018; Barrales-Cureño et al.,
same moieties with C–C bond formation at C-10 with C-16′ like in

4
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Table 2
Ethnomedicinal applications of different herbal preparations of plant parts of C. roseus in world-wide.
S. Plant Preparation Ethnomedicinal use Country Reference
N. part

1. Whole Decoction Muscle relieve pain, depression of CNS, wasps stings India Ross (2003); Don (1999) Das and Sharangi
plant (2017); Renjini et al. (2017); Senbagalakshmi
et al. (2017)
Decoction Nose bleed, bleeding gums, mouth ulcers, sore throats India Senbagalakshmi et al. (2017)
The hot water extract taken Cancer India Senbagalakshmi et al. (2017); Aslam et al.
orally (2010)
The plant was boiled to Stop bleeding Hawaii (Das and Sharangi, 2017; Renjini et al., 2017;
make a poultice Aslam et al., 2010)
Decoction Diabetes, cancer Senbagalakshmi et al. (2017)
Hot water extract of root Diabetes mellitus Brazil Senbagalakshmi et al. (2017); Aslam et al.
bark, dried entire plant (2010)
Hot water extract of dried Diabetes Thailand Senbagalakshmi et al. (2017); Aslam et al.
entire plant (2010)
Decoction Kidney problems, diabetes, arthritis Marshall Senbagalakshmi et al. (2017); Aslam et al.
Islands (2010)
Extract Cure stomach ulcer, sedative, and anticancer Japan Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Hot water extract of dried Antigalactagogue England Aslam et al. (2010)
entire plant
Juice For high blood pressure Dutch Senbagalakshmi et al. (2017)
Hot water extract of entire Antigalactogogue Vietnam Senbagalakshmi et al. (2017); Aslam et al.
plant (2010)
Hot water extract of entire Antigalactagogue France (Senbagalakshmi et al. (2017); Aslam et al.
plant (2010)
Decoction Used for diabetes Cuba (Das and Sharangi, 2017; Renjini et al., 2017;
Aslam et al., 2010)
Hot water extract of dried Cancer, heart disease, and leishmaniasis Peru Senbagalakshmi et al. (2017); Aslam et al.
entire plant (2010)
Decoction High blood pressure Venda Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Gargling with an infusion Relieve pain from a sore throat, laryngitis, and chest USA Das & Sharangi (2017); Aslam et al. (2010)
of the plant complaints and ease luncogestion inflammation
Extract Eye irritation and infection Caribbean Senbagalakshmi et al. (2017)
Decoction Diabetes and sedative Caribbean Senbagalakshmi et al. (2017)
Decoction Diabetes Myanmar Aslam et al. (2010)
decoction Diabetes Malaysia Das and Sharangi (2017)
Decoction of dried entire Diabetes mellitus, liver diseases Taiwan Das and Sharangi (2017)
plant
2. Aerial Infusion Orally for diabetes Kenya Senbagalakshmi et al. (2017); Aslam et al.
parts (2010)
Hot water extract Menstrual regulator Vietnam Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Hot water extract Menstrual regulator, malaria China Renjini et al. (2017); Aslam et al. (2010)
3. Stem Hot water extract Diabetes West Indies Das and Sharangi (2017); Renjini et al.
(2017); Aslam et al. (2010)
Decoction Dysmenorrheal Indonesia Senbagalakshmi et al. (2017); Aslam et al.
(2010)
4. Leaves Infusion Menorrhagia, Toothache, memory loss, blood circulation India Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Hot water extract of leaves Diabetes mellitus, amenorrhea, Philippines Das and Sharangi (2017); Renjini et al.
(2017); Aslam et al. (2010)
Decoction Regulating blood sugar, reducing toothaches, cardiovascular Sri Lanka Senbagalakshmi et al. (2017)
diseases, soothing insect bites, improving memory and
circulation, healing wounds
Hot water extract Diabetes England Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Decoction Vomitive France Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Hot water extract Combat primary inertia in childbirth; diabete Dominica (Senbagalakshmi et al., 2017; Aslam et al.,
2010)
Hot water extract Menorrhagia and diabetes. Australia (Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Expressed juice together Anemia. Myanmar Senbagalakshmi et al. (2017); Aslam et al.
with the same amount of (2010)
honey.
Crush fresh leaves Ulcers, inflammation Myanmar Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Decoction Traditionally used for diabetes Spain Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Decoction Astringent, diuretic, cough remedy China Renjini et al. (2017), Aslam et al. (2010)
Hot water extract Diabetes, rheumatism, Mozambique Senbagalakshmi et al. (2017); Aslam et al.
(2010)
juice Indigestion, dyspepsia Mauritius Das and Sharangi (2017); Aslam et al., 2010)
(continued on next page)

5
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Table 2 (continued )
S. Plant Preparation Ethnomedicinal use Country Reference
N. part

Decoction Menorrhagia, rheumatism Africa Das and Sharangi (2017)


Decoction Toothache, fever, skin diseases, diuretic. India Chandran & Suarsana et al. (2015)
5. Dried Hot water extract is taken Hodgkin’s disease India Aslam et al. (2010)
leaves orally
Decoction Diabetes, hypertension, cancer Cook Islands Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Hot water extract Diabetes Kenya Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Hot water extract Diabetes Guatemala Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Hot water extract Diabetes Jamaica Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Hot water extract Menorrhagia, diabetes Africa Renjini et al. (2017)
Decoction Diabetes mellitus Europe Senbagalakshmi et al. (2017); Aslam et al.
(2010)
6. Flower An extract of the flower Eyewash for the eyes of infants Rodriguez Senbagalakshmi et al. (2017); Aslam et al.
Islands (2010)
Extract Eye wash for the eyes of infants Jamaica Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Decoction Asthma, tuberculosis, flatulence Bahamas Das and Sharangi (2017); Renjini et al. (2017)
7. Dried Hot water extract Diabetes Pakistan Aslam et al. (2010)
ovules
8. Root The extract is taken orally Menorrhagia India Senbagalakshmi et al. (2017)
Decoction Tonic, stomachic India Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Hot water extract of dried Emmenagogue Philippines Das and Sharangi (2017)
root used orally
Hot water extract of root Abortion, emmenagogue, dysmenorrheal with scanty flow Philippines Das and Sharangi (2017); Renjini et al.
abortion (2017); Aslam et al. (2010)
Infusion Diabetes Mexico Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Decoction Purgative, vermifuge, depurative, hemostatic; toothache France Aslam et al. (2010)
remedy
Hot water extract Hypertensive, febrifuge Mozambique Aslam et al. (2010)
Decoction Vomitive, purgative, vermifugl, depurative, hemostatic and Madagascar Das and Sharangi (2017)
toothache remedies
9. Dried Decoction Fevers, malaria. Brazil Aslam et al. (2010)
root
Decoction Stomach problem Kenya Senbagalakshmi et al. (2017); Aslam et al.
(2010)
Water extract Venereal disease Venda Aslam et al. (2010)
10. Root Hot water extract Febrifug Australia Aslam et al. (2010)
bark

epidermis of C. roseus leaf. They are formed by the combination of 4.4. Flavonoids and phenolic acids
compounds 27 and 55 due to nucleophilic attack on indole moiety
(Palaniappan et al., 2020). Alkaloids 111–115 were produced due to A total of 34 flavonoids (146–179) and nine phenolic acids
substitution or addition of CH3, OH, H- on terpene moiety of cathar­ (180–188) have been identified from different plant parts of C. roseus
anthine (55) at C-20′ as well as N- on vindole moiety. Leurosine-Nb-­ (Fig. 5A-C). The quercetin-3-O-arabinoside (147), rutin (148), quer­
oxide (126), pseudovinblastinediol (116), deacetoxyvinblastine cetin-3-O-(2,6-di-O-rhamnosyl) galactoside (149), quercetin-3-O-(2,6-
(desacetoxyvinblastine) (118), leurosidine-N-oxide (124) and leur­ di-O-rhamnosyl)glucoside (150) and quercetin-3-O-(2,6-di-O-rhamno­
ocolornbine (132) have been reported to have additional OH of the syl-glucoside) (151) were regarded as hexoside derivatives of quercetin
catharanthine (55) moiety. 3′ ,4′ -Anhydrovinblastine (117) yielded due in hydro-alcoholic extracts of the leaves of this plant. Similarly,
to hydrogenation of terpene of the catharanthine moiety. 17-Deacetox­ kaempferol ( 151) and its glucosides, kaempferol-3-O-(6-O-rhamnosyl
yvinblastine N’b-oxide ( 119), 20′ -deoxyvinblastine N’b-oxide (120) glucoside (153), kaempferol-3-O-(2,6-di-O-rhamnosyl) galactoside
and leurosidine-N-oxide (124) formed due to oxidation of terpene on (154) and kaempferol-3-O-(2,6-di-O-rhamnosyl) glucoside (155) have
N-atom of the catharanthine moiety. Compound 121 is regarded as a identified in this plant by HPLC-MS/MS. Isorhamnetin-3-O-(2,6-di-O-
type of bisindole bisindole alkaloid formed due to the substitution of rhamnosyl) galactoside (156), isorhamnetin-3-O-(2,6-di-O-rhamnosyl-
CH3CO with NH2 in vindoline (27) moiety. Leurosine (125), deacetox­ galactoside (isomer) (157), isorhamnetin-3-O-(2,6-di-O-rhamnosyl)
yleurosine (desacetoxyleurosine) (128), 5′ -oxoleurosine (129), glucoside (158), isorhamnetin-3-O-(6-O-rhamnosyl) glucoside (159),
19′ -oxoleurosine (130) and vinleurosine (131) have been formed due to isorhamnetin-3-O-(6-O-rhamnosyl) galactoside (160) and isorhamnetin-
double bond terpene epoxidation of catharanthine moiety. Similarly, all 3-O-(6-O-rhamnosyl) hexoside (161) were characterized as hexosides of
bisindole alkaloids have been synthesized in different parts of C. roseus isorhamnetin, whereas, syringetin-3-O-robinobioside ( 162) was pre­
especially leaves and suspension cell cultures via oxidation, epoxidation sented as glucoside of syringetin. 3′ ,4′ -di-O-methylquercetin-7-O-
and dehydrogenation (Frasci et al., 2000; Jacobs et al., 2004; Yang et al., [(4′′ →3′′′ )-2′′′ ,6′′′ ,10′′′ ,14′′′ -tetramethylhexadec-13′′′ -ol-14′′′ -enyl]-β-D-
2011, Zhao et al., 2013; Zhang et al., 2013; Baig et al., 2021). glucopyranoside (163), 4′ -O-methylkaempferol-3-O-[(4′′ →3′′′ )-
2 ,6 ,10 ,14 -tetramethylhexadecan-13′′′ -olyl]-β-D-glucopyranoside
′′′ ′′′ ′′′ ′′′

(164), 3′ ,4′ -di-O-methylbutin-7-O-[(6′′ →1′′′ )-3′′′ ,11′′′ -dimethyl-7′′′ -


methylenedodeca-3′′′ ,10′′′ -dienyl]-β-D-glucopyranoside (165) and 4′ -O-

6
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Fig. 2. Biosynthetic pathways of different classes of Catharanthus indole alkaloids. TDC: tryptophan decarboxylase, LAMT: Loganic acid-O-methyltransferases, SLS:
secologanin synthase, STR: strictosidine synthase, GO: geissoschizine oxidase (Redox115), (Redox216), SAT: stemmadenine O-acetyltransferase, ASO: O-acetyl­
stemmadenine oxidase, GS: geissoschizine synthase, HL3: hydrolase 3, HL4: hydrolase 4. HL1: hydrolase 1, T16H: tabersonine 16-hydroxylase, T3O: tabersonine 3-
oxidase, 16OMT: 16-hydroxytabersonine 16-O-methyltransferase, T3R: tabersonine 3-reductase, NMT: 2,3-dihydro-3-hydroxytabersonine-N-methyltransferase, D4H:
desacetoxyvindoline 4-hydroxylase, DAT: deacetylvindoline 4-O-acetyltransferase.

methylbutin-7-O-[(6′′ →1′′′ )-3′′′ ,11′′′ -dimethyl-7′′′ -hydroxymethylenedo- malvidin-3-O-(6-O-p-coumaroyl) glucoside (179) hirsutidin-3-O-gluco­
decanyl]-β-D-glucopyranoside (166) were a special type of flavonoids side (176) and hirsutidin-3-O-(6-O-p-coumaroyl) glucoside (178) have
formed due to a combination of hexoside and terpene in the methanol been reported as hexoside of petunidin (171), malvidin (170) and hir­
extract of hairy roots. Petunidin-3-O-glucoside (174), petunidin-3-O-(6- sutidin (172), respectively. Pelargonidin (167), cyaniding (167), peo­
O-p-coumaroyl) glucoside (177), malvidin-3-O-glucoside (175), nidin (169), 170–172 and delphinidin ( 173) were considered as

7
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Table 3
Various classes of phytochemical from C. roseus.
S. Class Sub-class Compound Reference
N.

1 Alkaloids MIAs Vincadifformine Ramirez and García-Rubio (2003); Jacobs et al. (2004); Levac
2 Minovincinine et al. (2008); Arora et al. (2010); Pan et al. (2010); Soares et al.
3 Echitoveniline (2012); Wang et al. (2014); Wesołowska et al. (2016); Alam et al.
4 Echitovenidine (2017); Kumar et al. (2018a); Kumar et al. (2018b); Zhu et al.
5 Echitovenine (2018); da Silva e Silva et al. (2019); Koel et al. (2020)
6 Tabersonine
7 11-Hydroxytabersonine
8 11-Methoxytabersonine
9 Nor 4-hydroxytabersonine
10 16-methoxytabersonine
11 16-Hydroxytabersonine
12 19-Hydroxy-11-methoxytabersonine
13 19-Acetoxy-11-hydroxytabersonine
14 19-Acetoxy-11-methoxytabersonine
15 Minovincine
16 Lochnericine
17 19(S)-epimisiline
18 Lochnerinine
19 Horhammericine
20 Horhammerinine
21 Lochneridine
22 20-epi-Lochneridine
23 Akuammicine
24 Vinervine
25 Cathaphylline
26 Tubotaiwine (Dihydrocondylocarpine)
27 Vindoline
28 Vindorosine
29 Deacetylvindoline
30 Deacetylvindoline
31 4-O-deacetyl-N,-demethylvindolin
32 4-O-deacetylvindorosine
33 N,-demethyldeacetylvindorosine
34 16-O-Demethylvindoline
35 16-O,N,-bisdemethyl-4-O -deacetylvindolin
36 16-Methoxy-2,3-dihydro-3-hydroxytabersonine
37 Bannucine
38 Vindolidine
39 Vincoline
40 Kitraline
41 Dihydrovindoline
42 Venalstonine
43 Epivindolinine (vindolinine)
44 5(S)-Hydroxy-14,15-dihydrovindolinine
45 Vindolinine-Nb-oxide (9-Epivindolinine-Nb-oxide)
46 Venalstonidine
47 11-Methoxy-2,16-di-hydrotabersonine
48 2,3-Dihydrotabersonine
49 Cathovaline
50 Deacetylcathovaline
51 Hydroxycathovaline
52 Vinpocetine
53 Vincamine
54 Trichophylline
55 Catharanthine
56 Vincarodine
57 Pleiocarpamine
58 Coronaridine
59 Stemmadenine
60 Dehydrosecodine
61 Akuammiline
62 Desacetylakuammiline
63 10-Hydroxy-deacetylakuammiline
64 10-O-methylsarpagine (Lochnerine)
65 Sarpagine
66 Perivine
67 Pericyclivine
68 Vellosiminol
69 21-hydroxycyclolochnerine
70 10-Hydroxydeformodihydropseudoakuammigine 10-O-L-
arabinopyranoside
71 10-Hydroxycathafoline 10-O-larabinopyranoside
72 Ajmaline
(continued on next page)

8
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Table 3 (continued )
S. Class Sub-class Compound Reference
N.

73 10-Hydroxynortetraphyllicine
74 Norseredamine
75 Tetraphyllicine
76 Pseudoyohimbine
77 Yohimbine
78 Reserpine
79 3,4,5,6-tetradehydroyohimbine
80 Ajmalicine
81 3-Isoajmalicine
82 Akuammigine
83 Tetrahydroalstonine
84 19-Epiajmalicine
85 3-Iso-19-epi-ajmalicine
86 3-Epi-19-epiajmalicine
87 3-Epiajmalicine
88 Reserpiline
89 7-Hydroxyindolenineajmalicine
90 Alstonine
91 Cathenamine
92 Pseudoindoxyl ajmalicine
93 Strictosidine lactam
94 Mitraphylline
95 Serpentine
96 Vincoside
97 Dihydrositsirikine
98 Sitsirikine
99 Isositsirikine (16R,19E)
100 Isositsirikine (16R,19Z)
101 O-Acetylstemmadenin (strychnos)
102 Vallesiachotamine
103 17-Isovallesiachotamine
104 Geissoschizine
105 Strictosidine (3α (S)-strictosidine)
106 Anthirine
107 β-Carboline
108 n,n-dimethyltryptamine
109 Tryptamine,Nb-acetyl
110 Iochrovicine
111 Bisindole Vinblastine Frasci et al. (2000); Jacobs et al. (2004); Yang et al. (2011), Wang
alkaloid et al. (2012a); Wang et al. (2012b); Zhao et al. (2013); Zhang et al.
112 Vincristine (2013); Alam et al. (2017); Jeong and Lim (2018); Kumar et al.
113 4′ -Deoxyvinblastine (2018a); Kumar et al. (2018b)
114 Demethylvinblastine
115 Deoxyvincaleukoblastine B
116 Pseudovinblastinediol
117 3′ ,4′ -Anhydrovinblastine
118 Deacetoxyvinblastine (Desacetoxyvinblastine)
119 17-Deacetoxyvinblastine N’b-oxide
120 20′ -Deoxyvinblastine N’b-oxide
121 Vindesine
122 Catharine
123 Leurosidine
124 Leurosidine-N-oxide
125 Leurosine
126 Leurosine-Nb-oxide (Pleurosine)
127 Isoleurosine
128 Deacetoxyleurosine (Desacetoxyleurosine)
129 5′ -Oxoleurosine
130 19′ -Oxoleurosine
131 Vinleurosine
132 Leurocolornbine
133 Roseadine
134 Vincathicine
135 Catharinine (Vinamidine)
136 Vincovaline
137 Vincovalinine
138 Vincovalicine
139 Vincadioline
140 Vindolicine
141 Catharanthamin
142 Vingramine
143 Vinorelbine
144 Vinrosidine
145 Methylvingramine
146 Flavonoid Flavonol Quercetin
(continued on next page)

9
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Table 3 (continued )
S. Class Sub-class Compound Reference
N.

147 Quercetin-3-O-arabinoside Piovan and FilippiniFavretto (1998); Kessler et al. (2003); Choi
148 Rutin et al. (2002); Piovan and Filippini (2007); Yang et al. (2008);
149 Quercetin-3-O-(2,6-di-O-rhamnosyl) galactoside Chung et al. (2009); He et al. (2010); Ferreres et al. (2011); Rao
150 Quercetin-3-O-(2,6-di-O-rhamnosyl) glucoside and Ahmed (2014); Nisar (2017a); Nisar (2017b); Lahare et al.
151 Quercetin-3-O-(2,6-di-O-rhamnosyl-glucoside) (2020); Yu et al. (2021)
152 Kaempferol
153 Kaempferol-3-O-(6-O-rhamnosyl) glucoside
154 Kaempferol-3-O-(2,6-di-O-rhamnosyl) galactoside
155 Kaempferol-3-O-(2,6-di-O-rhamnosyl) glucoside
156 Isorhamnetin-3-O-(2,6-di-O-rhamnosyl) galactoside
157 Isorhamnetin-3-O-(2,6-di-O-rhamnosyl) galactoside (isomer)
158 Isorhamnetin-3-O-(2,6-di-O-rhamnosyl) glucoside
159 Isorhamnetin-3-O-(6-O-rhamnosyl)glucoside
160 Isorhamnetin-3-O-(6-O-rhamnosyl) galactoside
161 Isorhamnetin-3-O-(6-O-rhamnosyl) hexoside
162 Syringetin-3-O-robinobioside
163 3′ ,4′ -di-O-methylquercetin-7-O-[(4′ ′ →3′′′ )-2′′′ ,6′′′ ,10′′′ ,14′′′ -
tetramethylhexadec-13′′′ -ol-14′′′ -enyl]-β-D-glucopyranoside
164 4′ -O-methylkaempferol-3-O-[(4′ ′ →3′′′ )-2′′′ ,6′′′ ,10′′′ ,14′′′ -
tetramethylhexadecan-13′′′ -olyl]-β-D-glucopyranoside
165 3′ ,4′ -di-O-methylbutin-7-O-[(6′ ′ →1′′′ )-3′′′ ,11′′′ -dimethyl-7′′′ -
methylenedodeca-3′′′ ,10′′′ -dienyl]-β-D-glucopyranoside
166 4′ -O-methylbutin-7-O-[(6′ ′ →1′′′ )-3′′′ ,11′′′ -dimethyl-7′′′ -
hydroxymethylenedo-decanyl]-β-D-glucopyranoside
167 Anthocyanin Pelargonidin
168 Cyanidin
169 Peonidin
170 Malvidin
171 Petunidin
172 Hirsutidin
173 Delphinidin
174 Petunidin-3-O-glucoside
175 Malvidin-3-O-glucoside
176 Hirsutidin-3-O-glucoside
177 Petunidin-3-O-(6-O-p-coumaroyl) glucoside
178 Hirsutidin-3-O-(6-O-p-coumaroyl) glucoside
179 Malvidin-3-O-(6-O-p-coumaroyl) glucoside
180 Phenolic Phenolic acid Gallic acid Kroes et al. (1992); Kratz et al. (2008); Li et al. (2020); Nisar
acid (2017a)
181 Ferulic acid
182 Caffeic acid
183 Benzoic acid
184 Vanillic acid
185 Coniferyl aldehyde
186 4-O-Caffeoylquinic acid
187 5-O-Caffeoylquinic acid
188 3-O-Caffeoylquinic acid
189 Volatile Alkane n-Dodecane (Brun et al. (2001); Pandey-Rai et al. (2006); De Pinho et al.
compound (2009); Lawal et al. (2015); Fitrianto et al. (2020); Syeda and
190 n-Tridecane Riazunnisa (2020)
191 n-Tetradecane
192 n-Pentadecane
193 Heptadecane
194 Octadecane
195 Nonadecane
196 Eicosane
197 Heneicosane
198 2,6,11-Trimethyl dodecane
199 Docosane
200 Tricosane
201 Tetracosane
202 Dotriacontane
203 Tetracontane
204 Alkene n-Hexadecene
205 Heneicosene
206 Docosene
207 (Z)-9-Tricosene
208 (Z)-β-Ocimene
209 Myrcene
210 (E,E)-α-Farnesene
211 3-Phenyldodecane
212 Limonene
213 β-Phellandrene
214 γ-Terpinene
215 β-Bisabolene
(continued on next page)

10
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Table 3 (continued )
S. Class Sub-class Compound Reference
N.

216 Germacrene D
217 γ-Cadinene
218 Trans-α-Bergamotene
219 α-Pinene
220 β-Pinene
221 Aldehyde Heptanal
222 Octanal
223 Nonanal
224 Decanal
225 Undecanal
226 Dodecanal
227 Tetradecanal
228 Pentadecanal
229 Hexadecanal
230 (E)-2-Octenal
231 (E)-2-Nonenal
232 (E)-2-Decenal
233 (E)-2-Undecenal
234 (E,E)-2,4-Hexadienal
235 (E,E)-2,4-Heptadienal
236 (E,E)-2,4-Decadienal
237 (E,E)-2,4-Dodecadienal
238 (E,Z)-2,4-Decadienal
239 (E, Z)-2,6-Nonadienal
240 Neral
241 6,6-Trimethylcyclohexen-1-acetaldehyde
242 Perilla aldehyde
243 β-Cyclocitra
244 Safranal
245 Benzaldehyde
246 4-Methylbenzaldehyde
247 Ethylbenzaldehyde
248 Ketone 2-Methyl 4-heptanone
249 Pentadecane-2-one
250 Hexahydrofarnesylacetone
251 Heptenone
252 3-Octen-2-one
253 Isophorone
254 β-Pulegone
255 (E)-Ionone-5,6-epoxide
256 (E)-α-Ionone
257 (E,E)-Octa-3,5-dien-2-one
258 (E,Z)-Octa-3,5-dien-2-one
259 6-Methyl-3,5-heptadien-2-one
260 (E)-β-Ionone
261 (E,E)-Pseudoionone
262 Megastigmatrienone
263 (E)-Damascenone
264 2,6-bis(1,1-dimethylethyl)- 2,5-Cyclohexadiene-1,4-dione
265 6-Methyl hept-5-ene-2-one
266 2,2,6-Trimethylcyclohexanone
267 6-Methyl-5-hepten-2-one
268 Geranylacetone
269 Farnesylacetone
270 Davanone
271 Alcohol Dodecyl alcohol
272 Hexadecanol
273 Eicosan-1-ol
274 Phytol
275 Citronellol
276 9-Octadecen-1-ol
277 Nerol
278 Geraniol
279 (E,E)-Farnesol
280 (Z,Z)-Farnesol
281 (E,Z)-Farnesol
282 Iso-Dihydro carveol
283 Isopulegol
284 Linalool
285 Isophytol
286 1-[(E)-1-Hexenyl]-cyclohexanol
287 Trans-Sabinene hydrate
288 α-Eudesmol
289 γ-Eudesmol
290 Terpinen-4-ol
(continued on next page)

11
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Table 3 (continued )
S. Class Sub-class Compound Reference
N.

291 α-Terpineol
292 Cedrol
293 Widdrol
294 Elemol
295 2,6-di-tert -butyl-p-cresol
296 2-Hydroxy 5-methoxy p-cymene
297 (E)-Anethole
298 Eugenol
299 Methyl eugenol
300 Methyl salicylate
301 Lanosterol
302 β-Sitosterol
303 α-Amyrin
304 Acid Caproic acid
305 Caprylic acid
306 Nonanoic acid
307 Capric acid
308 Undecanoic acid
309 Lauric acid
310 Tridecanoic acid
311 Myristic acid
312 Pentadecanoic acid
313 Palmitic acid
314 Stearic acid
315 Hexadecenoic acid
316 Linoleic acid
317 Octadecenoic acid
318 Dehydroabietic acid
319 Methyl heptanoate
320 Methyl caprylate
321 Methyl undecanoate
322 Methyl myristate
323 Methyl pentadecanoate
324 Methyl palmitate
325 Methyl 9-tridecenoate
326 Hexatrienoic acid methyl ester
327 Linoleic acid methyl ester
328 Linolenic acid methyl ester
329 Oleic acid ethyl ester
330 Linolenic acid ethyl ester
331 Isoamyl laurate
332 Ester Dodecyl acetate
333 Hexadecyl acetate
334 Neryl acetate
335 (E,E)-Farnesyl acetate
336 α-Terpinyl acetate
337 α-Tocopherol
338 Acetate Dihydroactinidiolide
339 (Z)-Linalool oxide
340 (E)-Linalool oxide
341 Theaspirane B
342 Theaspirane A
343 Edulane II
344 1,8-Cineole

anthocyanins and formed due to the substitution of hydrogen from O–H especially leaves of C. roseus (Brun et al., 2001; Pandey-Rai et al., 2006;
with an alkyl group. Gallic acid (180), ferulic acid ( 181), caffeic acid De Pinho et al., 2009; Lawal et al., 2015; Fitrianto et al., 2020; Syeda and
(182), benzoic acid (183), vanillic acid (184), coniferyl aldehyde (185), Riazunnisa, 2020). All these volatile compounds reported from C. roseus
4-O-caffeoylquinic acid (186), 5-O-caffeoylquinic acid ( 187) and 3-O- are presented in Fig. 6A and B.
caffeoylquinic acid (188) have been identified and characterized as
phenolic acids especially gallic and caffeic acid derivatives from the 5. Quality control/quality assurance: separation and analysis
different extracts of leaves (Kessler et al., 2003; Piovan and Filippini, techniques
2007; Yang et al., 2008; Ferreres et al., 2011; Rao and Ahmed, 2014;
Nisar, 2017a, 2017b). The separation and analysis techniques such as thin-layer chroma­
tography (TLC), high-performance thin-layer chromatography (HPTLC),
high-performance liquid chromatography (HPLC), HPLC-MS, HPLC-MS/
4.5. Volatile compounds MS, gas chromatography (GC) and GC-MS have been evaluated herbal
products for quality control (Kumar et al., 2016a, 2016b). Several
Total of 156 volatile compounds including alkanes (189–203), al­ analytical methods have been reported for separation, qualitatitation
kenes (204–220), aldehydes (221–247), ketones (248–270), alcohols and characterization of bioactive compounds from complex matrix such
(271–304), acids (305–319), esters (320–332), acetates (333–338), plant extracts mainly ethanolic and methanolic as well as alkaloidal
lactones (339–344) were summarized from different plant parts

12
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Fig. 3A. Structures of monoterpene indole alkaloids (1–59) from C. roseus.

fractions of plant parts of C. roseus to speed up drug discovery process 1985; Naaranlahti et al., 1987; Sim et al., 1994; Chu et al., 1997; Uniyal
and quality control/quality assurance. The analytical methods and et al., 2001; Zhao et al., 2001; Choi et al., 2002).
separation techniques and their methodology are presented in Table 4.
The analytical approaches classified as (i) qualitative analysis which
aims metabolic profiling, to display a large number of primary and 5.1. Analysis of monoterpene indole alkaloids
secondary metabolites from the extracts, (ii) quantitative analysis of
targeted phytochemicals from complex matrix (Tang et al., 2020). Most 5.1.1. Qualitative analysis
of the qualitative and quantitative HPLC and HPLC-MS methods have Mostly of the qualitative analysis of MIAs has been reported on C18
been reported which are made up of modification of previously reported column for separation with the mobile phase of a combination of
analytical methods using C18 column and some time C8 also used with acetonitrile and methanol with a buffer like formic acid with isocratic or
different buffer systems as a mobile phase. All HPLC-MS methods have gradient systems (Renaudin 1984, 1985; Bhadra and VaniShanks, 1993;
been reported in positive ionization mode for MIAs (Renaudin 1984, Moreno-Valenzuela et al., 1998; Rijhwani and Shanks, 1998; Lee and
Shuler, 2000; El-Sayed et al., 2004). A combination of different type of

13
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Fig. 3B. Structures of monoterpene indole alkaloids (60–110) from C. roseus.

buffer solutions such as ammonium carbonate, ammonium acetate, formic acid was preferred for HPLC-MS analysis due to avoid suppres­
ammonium formate and sodium phosphate buffer have been also used sion of ions during analysis while HPLC allow for all types of buffer
adequately as mobile phase for separation of MIAs by the many different systems as mobile phase (Konermann, 2017 Susa et al., 2017). Many
research groups (Auriola et al., 1989; Toivonen et al., 1991; Schröder research groups have identified and characterized compounds 27, 80,
et al., 1999; Singh et al., 2000; Barthe et al., 2002; Tikhomiroff and 16-O-demethylvindoline (34), and serpentine (95) in alkaloid extracts of
Jolicoeur, 2002; Batra et al., 2004; Lee-Parsons et al., 2004; Dutta et al., different parts of C. roseus using LC-MS (Chu et al., 1997; Choi et al.,
2005; Shams et al., 2009; Chen et al., 2013; Koel et al., 2020; Baig et al., 2002; Zhou et al., 2005; He et al., 2011; Chen et al., 2013; Kumar et al.,
2021). A different ratio of acetonitrile and methanol with a buffer like 2018a, 2018b; Jeong et al., 2018). Similarly, compounds 27, 34, 95,

14
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Fig. 4A. Structures of bisindole alkaloids (111–133) from C. roseus.

15
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Fig. 4B. Structures of bisindole alkaloids (134–145) from C. roseus.

111, 112, and vindolidine (38), vindolinine (43) and 19-S-vindolinine aqueous extracts of C. roseus.Thermo Betasil C8 column (250 × 4.5 mm,
(isomer of vindolinine) have been identified and characterized from the 5 μm) at 25 ◦ C used for chromatographic separation of MIAs using
leaves of an alkaloids extract using high-performance liquid mobile phase, 0.1% formic acid/water, and acetonitrile using
chromatography-electrospray ionization tandem mass spectrometry high-performance liquid chromatography (HPLC) coupled with elec­
(HPLC-ESI-MS/MS) (Rischer et al., 2006; Chen et al., 2011; Chen et al., trospray ionization quadrupole time of flight tandem mass spectrometry
2013; El-Domyati et al., 2014; El-Domyati et al., 2014; Ayob et al., (HPLC-ESI-QTOF-MS/MS) in positive mode (Kumar et al., 2018a).
2017). A total of 16 alkaloids have been identified and characterized in Compounds 27, 52, 34, 111, 112 and 121 have been unambiguously
extracts of whole stem tissue by HPLC-ESI-MS/MS (Luo et al., 2014; identified and characterized using diagnostic fragmentation pathways
Yamamoto et al., 2016). The HPLC-ESI-MS/MS method has been (Kumar et al., 2018a). A total of 72 MIAs alkaloids which included 11
developed for the characterization of compounds 27, 34, 111, 112 and MIAs were unambiguously identified and characterized by comparison
131 by comparison with the standards in three batches of C. roseus crude with standards, whereas the remaining 62 MIAs were tentatively iden­
samples collected from different regions of China. A capillary electro­ tified and characterized in ethanolic extracts of leaves, stem and roots of
phoresis–mass spectrometry (CE-MS) method has been used for the the plant. The method has been very useful for the separation and
identification of compounds 111 and its monomeric precursors ( 27) and identification of isomers of well known compounds. Distribution of all
compound 34 from stems of C. roseus (Chen et al., 2011). Compounds the MIAs have been investigated in 30 samples of C. roseus collected
27, 34, 80, 111 and 131 were identified from stems of C. roseus using from five states of India. The approach considered as rapidly and
direct-injection in ion trap-mass spectrometry (IT-MS) (Chen et al., effectively characterizes the targeted and untargeted MIAs in C. roseus
2013). using developed analytical methods together with diagnostic fragment
The ultra-performance liquid chromatography-quadrupole time-of- patterns (Kumar et al., 2018a). Total 172 alkaloids mass spectrometric
flight (UPLCQ-TOF) mass spectrometry method has been developed for data for dereplication along with computer-based approaches, such as
the identification and characterization of compounds 27, 34, 80, 95, molecular networking has been reported which are the maximum
111 and 112. A total of 22 indole alkaloids, including, compounds 27, number of MIAs identified in this plant till date (Ramos et al., 2019).
34, 80, 95, 55, 111 and 112 have been unambiguously characterized, Relatively advanced approach 13carbon (13C) and 15nitrogen (N) label­
whereas around 16 compounds were tentatively identified (Jeong and ing along molecular networkinghas with applied on HPLC-MS/MS data
Lim, 2018). Ferreres et al. (2010) have been reported an by Nakabayashi et al. (2020). The characteristics of two types of skel­
HPLC-ESI-MS/MS for identification and characterization of compounds eton namely indole basic and oxindole skeletons have been have pro­
43, 34 and 80 along with isomers of compounds 43, 80 and 95 in posed on the basis of mass (m/z) shifting of the product and precursor

16
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Fig. 5A. Structures of flavonoids (146–166) from C. roseus.

ions. Total of 86 and 73 common monoisotopic ions of indole basic and pharmaceutical industries. Like qualitative analysis, numerous methods
oxindole skeletons reported, respectively. The comparative analyses of have been reported for the quantitation of compounds 111 and 112
the three pairs of stereoisomers showed more than 170 common mon­ along with different alkaloids using both C18 and C8 column with buffer
oisotopic ions in each pair (Nakabayashi et al., 2015). Finally, the mo­ system as mobile phase (BlaskÓ and Cordell, 1990; Favretto et al.,
lecular formula of 45 MIAs was unambiguously identified using isotopic 2001). These compounds have been quantified from ethanolic extract
15
N. The alkaloid network analysis showed the structural commonality using TLC (Tam et al., 1995). Compounds 111 and 112 have been
and uniqueness among the MIAs. Some of them, antirhine was identified typically quantified using TLC and HPTLC. Similarly, quantitation of
using an authentic standard in leaves of this plant (Nakabayashi et al., compounds 111 and 112 has been reported by TLC and HPLC and
2020). Serpentinine-related bisindole alkaloids have been characterized ultraviolet–visible (UV–Vis), HPLC-ESI-MS, and HPLC-ESI-MS/MS
using using LC-MS/MS and NMR guided by a molecular (Toivonen et al., 1991; Zhang et al., 2017). Another bisindole alka­
networking-based dereplication strategy in ethanolic extracts Picralima loid, isoleurosine has been quantified using UV and TLC (Mon­
nitida (Alcover et al., 2020). The investigation of published literature forte-González et al., 1992; Nagy-Turak and Vegh, 1994; Parthasarathy
arrive at a conclusion that the MS/MS analysis with isotopic labelling et al., 2020). Similarly, detection of various bisindole alkaloids from
along with computer based networking analysis provides the confidence extracts of C. roseus and its cell culture have been reported by HPLC
of dereplication MIAs in complex metrix. coupled with different detectors such as photo diode array (PDA),
fluorescence and mass spectrometry (MS) for quantitative analysis
5.1.2. Quantitative analysis (Wittkamp et al., 1997; Gupta et al., 2005; Kumar et al., 2013;
The large-scale quantification of compounds 111 and 112 from Thompson, 2017; Thiäner et al., 2019).
different extracts of plant parts of C. roseus made special attention to Several improvements and developments were made to make HPLC

17
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Fig. 5B. Structures of pronthocynine (167–179) and phenolic acids (180–188) from C. roseus.

an even more powerful analytical technique and enhance the capacity source using an IT-MS instrument in the positive ion mode (Zhou et al.,
via integrated more powerful detectors such as mass spectrometry (MS). 2005; Kumar et al., 2018b). These HPLC-MS/MS methods showed good
The measurement of molecular weight for each peak is considered signals corresponding to the protonated molecular ions [M+H]+ and
highly useful to analyze plant or cell extracts by liquid chromatogra­ product ions (Zhang et al., 2017). The MS combined with ESI provided a
phy–mass spectrometry (LC-MS). A method for quantitative analysis of high selectivity and sensitivity for the determination of MIAs in positive
MIAs in plant extracts has been reported by Van der Heijden et al. (1987) mode. The separation, identification and quantification of MIAs after
using mass spectrometry for the first time. The HPLC coupled with validation of the developed methods were applied in extracts of C. roseus
electrospray ionization (ESI) ion source enhanced the detection of using HPLC with UV and MS detection. The amount of compounds 111
compounds 112 and 111 in plant extracts at a limit of detection of 0.2 and 131 observed were 1.8, 2.10, 6.02 and 7.52 μg/g by LC–UV and
μg/mL. The sensitivity for MIAs was 2–3 times higher in ESI-MS rather LC-MS, respectively (Chen et al., 2013). Compounds 111, 112 and 131
than with UV detection (Chu et al., 1997; Zhao et al., 1999). Most of the were separated on a C18 HPLC column with the mobile phase of meth­
quantitative HPLC and LC-MS methods have been validated according to anol with buffer 15 nmol/L ammonium acetate with 0.02% formic acid
the ICH guidelines. using multiple reaction monitoring (MRM) mode. This method was
Due to the advancement of analytical instruments, compounds 111 validated and the results observed as the intra- and inter-day precision
and 131 have been identified in C. roseus by direct infusion into ESI and accuracy of these alkaloids were within 1.2–11.5 (RSD: relative

18
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Fig. 6A. Structures of volatile compounds (alkanes (189–203), alkenes (204–220), aldehydes (221–247), ketones (248–270) from C. roseus.

19
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Fig. 6B. Structures of volatile compounds (alcohols (271–303), acids (304–318), esters (319–331), acetates (332–337), lactones (338–344) from C. roseus.

20
S. Kumar et al.
Table 4
Separation and analytical techniques and their conditions used for analysis of Catharanthus alkaloids in different extracts.
S.N. Instrument Analytes Column Mobile Phase Detection Elution Range of RT Flow Plant part Extract Reference
program (min) rate
(mL/
min)

1. HPLC Ajmalicine, μBondapack C18 Methanol:5 mM (NH4)2HPO4 (pH UV254 Isocratic 12.00–19.00 1 Cell Ethanolic Renaudin (1984)
catharanthine, (300 × 3.9 mm, 7.3) (71:29, v:v) suspension
serpentine and 10 μm) (Waters) cultures
vinblastine
Ajmalicine, μBondapack C18 Methanol:5 mM (NH4)2HPO4 (pH UV254 Isocratic 9.5–22 1 Hairy roots, Aqueous solution Renaudin (1985)
catharanthine, (300 × 3.9 mm, 7.3) (67:33, v:v) cell with 001% acetic
tetrahydroalstonine 10 μm) (Waters) suspensions acid
and vindoline
Ajmalicine, Spheri-5 C18 (220 Solvent A: 0.2% TEA in 25 mM PDA Gradient 13–24.5 Cell Ethanolic Naaranlahti et al.
catharanthine, × 4.6 mm, 5 μm) CH3COONH4; Solvent B: 0.2% TEA suspension (1987)
serpentine, in a mix of methanol: acetonitrile cultures
vinblastine, (29:71, v:v)
vincristine and
vindoline
Ajmalicine, μBondapak C18 Acetonitrile: 0.1 M CH3COONH4 UV280 Gradient 2.0–18.0 1 Tissue Ethanolic Auriola et al. (1989)
catharanthine, (300 × 3.9 mm, (pH 7.2) (49:51, v:v) cultures
serpentine, 10 μm)
tabersonine and
tryptamine
Catharanthine and Supelcosil LC-18- 0.1 M sodium acetate: methanol: UV 0–20 1 Catharanthus ethanol Naaranlahti et al.
ajmalicine DB (150 × 4.6 acetonitrile (49: 15: 36, pH 6.5) roseus cell (1989)
mm, 5 μm) culture
21

Ajmalicine, Hypersil ODS C18 Solvent A: 14 mM TEA in PDA Gradient N/A 0.4 Hairy root Ethanolic Toivonen et al.
catharanthine and (100 × 2.1 mm, 5 CH3COONH4; Solvent B: 14 mM cultures (1991)
tabersonine μm) (Agilent) TEA in Methanol: acetonitrile
(50:50, v:v)
Ajmalicine, μBondapack C18 Methanol:5 mM (NH4)2HPO4 (pH UV254 Isocratic 8.5–59.20 0.8 Hairy roots, alkaloidal Bhadra and
catharanthine, (300 × 3.9 mm, not mentioned) (67:33, v:v) cell VaniShanks (1993)
serpentine, 10 μm) (Waters) suspensions
vinblastine and
vindoline
Catharanthine, Hypersil ODS Solvent A: Water, 0.1% TEA; PDA Gradient N/A Cell Ethanolic Aerts et al. (1994)
tabersonine and (200 × 2.1 mm, 5 Solvent B: Acetonitrile, 0.1% TEA suspension
vindoline μm) (Agilent) cultures
Ajmalicine and μBondapack C18 Methanol: acetonitrile: 5 mM UV254 Isocratic NA 1 Cell alkaloidal Sim et al. (1994)
catharanthine (300 × 3.9 mm, (NH4)2HPO4(pH 7.3) (30:40:30, v: suspension
10 μm) (Waters) v:v) cultures

Journal of Ethnopharmacology 284 (2022) 114647


16- Nova-Pak C18 Methanol: water: TEA (75:25:0.1, v: PDA Gradient 30.5 0.6 Cell Crude protein St-Pierre and De
methoxytabersonine (300 × 3.9 mm, 4 v:v) suspension Luca (1995)
μm) (Millipore) cultures
Ajmalicine and Ultrasphere ODS Acetonitrile:10 mM (NH4)2HPO4, UV280 Isocratic 5.9–21.80 1.5 Hairy roots, Not reported Moreno-Valenzuela
yohimbine C18 (250 × 4.6 (43:57, v:v) cell et al. (1998)
mm, 4 μm) suspensions
(Beckman)
Ajmalicine, Bondclone C18 Methanol: acetonitrile: 5 mM PDA Isocratic NA 1 Cell alkaloidal Rijhwani and
hörhammericine, (300 × 3.9 mm, (NH4)2HPO4 (32:32:36, v:v:v) suspension Shanks (1998)
lochnericine, 10 μm) cultures
serpentine and (Phenomenex)
tabersonine
16- Solvent A:0.2% NaOH in water, PDA Gradient 13.0–26.7 1 Schröder al.et.
hydroxytabersonine, Solvent B: Acetonitrile (1999)
(continued on next page)
Table 4 (continued )

S. Kumar et al.
S.N. Instrument Analytes Column Mobile Phase Detection Elution Range of RT Flow Plant part Extract Reference
program (min) rate
(mL/
min)

16-methoxytaberso­ Nucleosil C18


nine and tabersonine (200 × 4 mm, 5
μm) (Agilent)
Catharanthine, μBondapack C18 Acetonitrile:0.1 M Phosphate PDA Isocratic 10.15–13.54, 0.6 Leaves 90% ethanolic Singh et al. (2000)
vinblastine, vindoline (300 × 3.9 mm, buffer: glacial acetic acid (pH 4.14)
and vincristine 10 μm) (Waters) (38:62:0.3, v:v:v)
Catharanthine, Symmetry C18 Solvent A (pH 7.0): Methanol: PDA Gradient 14.62–19.6 Cell – Uniyal et al.,(2001)
vinblastine (150 × 4 mm, 5 acetonitrile:5 mM CH3COONH4: suspension
Vincristine and μm) (Waters) triethylamine (13:32:55:0.2, by cultures and
vindoline volume); Solvent B (pH 7.0): plant parts
Methanol: acetonitrile:5 mM
CH3COONH4: triethylamine
(19:46:35:0.2, by volume)
Ajmalicine, Nucleosil 5 C18 Methanol: acetonitrile: 25 mM PDA Gradient N/A 1 Cell Methanolic Zhao et al. (2001)
catharanthine and (250 × 4.6 mm, 5 CH3COONH4: TEA (15:40:45:0.1, suspension
serpentine μm) (Agilent) by volume) cultures
Anhydrovinblastine, XTerra C18 (250 Acetonitrile: water: boric acid (pH UV214 Isocratic 6.0–20.8 1 – – Barthe et al. (2002)
catharanthine, × 4.6 mm, 5 μm) 10) (55:45:3.1, v: v: w) nm
vindoline, vinflunine (Waters)
and viorelbine
Catharanthine, Zorbax XDB C18 Solvent A: 5 mM KH2PO4 (pH 6); UV Gradient 7.8–20.8 2 Hairy root Tikhomiroff and
serpentine, (250 × 4.6 mm, 5 Solvent B: Acetonitrile cultures Jolicoeur (2002)
tabersonine, μm) (Agilent)
vinblastine, vindoline
and vincristine
22

Ajmalicine and μBondapack C18 Acetonitrile:0.1 M Phosphate UV Isocratic 0.6 Root 90% ethanolic Batra et al. (2004)
serpentine (300 × 3.9 mm, buffer:glacial acetic acid (pH 4.14)
10 μm) (Waters) (38:62:0.3, v:v:v)
Ajmalicine, Phenomenex DAD NA Leaf and root Digvijay et al.
catharanthine, Luna C18 (250 × (2004)
loganine, serpentine, 4.6 mm, 5 μm)
tryptamine,
tryptophan,
vinblastine,
vincristine and
vindoline
Ajmalicine, C18 (125 × 4 mm, 0.025 M UV233, Gradient – 1 Leaves, stems, Methanolic Favali et al. (2004)
serpentine, 5 μm) methanol–acetonitrile–ammonium 254, 280 and roots
vinblastine and acetate

Journal of Ethnopharmacology 284 (2022) 114647


vindoline
Catharanthine, Vydac C18 (250 × Acetonitrile:0.1% (w:v) TFA in UV254 Isocratic 7.2–23.5 1 Cell alkaloidal El-Sayed et al.
stemmadenine and 4.6 mm, 5 μm) water (21:79, v:v) suspension (2004)
tabersonine (Vydac) cultures
Ajmalicine, μBondapack C18 Acetonitrile: 0.1% (w:v) UV254 Isocratic 19.67–23.14 1 Cell – Lee-Parsons et al.
catharanthine, (300 × 3.9 mm trifluoroacetic acid (TFA) in water suspension (2004)
serpentine 10 μm) (Waters) (22:78, v:v) cultures
Ajmalicine, Phenomenex 12% acetonitrile: 88% water + UV254 Isocratic May-37 1 Cell Methanolic Wong et al. (2004)
serpentine and Luna C18 (150 × 0.1% formic acid suspension
tryptamine 4.6 mm, 5 μm) cultures
Ajmalicine, Waters’ıBondapk CH3OH: 5 mM NH4H2PO4(7: 3, v/v, UV – 1 Suspension Methanolic Zheng &Wu (2004)
tryptamine and C18 (250 × 4.6 pH 7.1) culture of the
tryptophan mm, 5 μm) cell line
UV Gradient 2 Leaves 90% ethanolic Dutta et al. (2005)
(continued on next page)
S. Kumar et al.
Table 4 (continued )
S.N. Instrument Analytes Column Mobile Phase Detection Elution Range of RT Flow Plant part Extract Reference
program (min) rate
(mL/
min)

Catharanthine, Zorbax XDB C18 Solvent A: 5 mM KH2PO4 (pH 6);


serpentine, (250 × 4.6 mm, 5 Solvent B: Acetonitrile
tabersonine, μm (Agilent)
vindoline, vincristine
and vinblastine
Catharanthine, Chromolith 21:79 (v/v) acetonitrile–0.1M UV254 Gradient 6.81–26.93 1.2 Leaves 90% Ethanolic Gupta et al. (2005)
vinblastine, Performance C18 phosphate buffer containing 0.5%
Vincristine and (100 × 4.6 mm) glacial acetic acid
vindoline
Ajmalicine, Luna C18 (150 × Solvent A: Water, Solvent B: UV Gradient 5.0–37.0 1 Hairy root Lee-Parsons et al.
serpentine and 4.6 mm 5 μm) Acetonitrile, Solvent C: Formic acid cultures (2004)
tryptamine (Phenomenex) (kept at 0.1% of the total elution
volume)
Catharanthine, Inertsil ODS-3 C18 5 mM sodium phosphate pH 6.0 UV220,230 Gradient 2 Leaves alkaloidal Murata and Luca
tabersonine and (4 × 250 mm) (solvent A) and acetonitrile (solvent (2005)
vindoline B)
Vinblastine, Agilent Zorbax 0.1% Triethylamine (solvent A) and UV280 Gradient 10.0–40.0 0.8 Leaves Commercial Zhou et al. (2005)
vincristine, vindoline Eclipse XDB-C8 methanol (solvent B)
and vindolidine (150 × 4.6 mm, 5
μm)
Ajmalicine, Phenomenex 50% MeOH/50% MeCN: 5 mM UV218 Gradient 1 Hairy roots Methanolic and Peebles et al. (2006)
catharanthine, Luna 5A C18 (250 (NH4)2HPO4 (pH 7.3) alkaloidal
horhammericine, × 4.6 mm)
lochnericine, (Torrance, CA)
23

serpentine,
tabersonine,
tryptamine and
tryptophan
Catharanthine, ODS C18 (250 × water + 0.1% Formic acid and B UV220 – Leaves and Methanolic Guo et al. (2007)
vinblastine, vindoline 4.6 mm) (Waters) acetonitrile + 0.1% formic acid roots
and vincristine
Horhammericine BEH C18 (1.0 × A: methanol: acetonitrile:5 mM: UV Isocratic – Hairy root alkaloidal Magnotta et al.
50 mm 1.7 μm NH4OAc:Et3N (5.8:14.2:80:0.2 by cultures (2007)
mm) (Waters, volume) B: MeOH:MeCN:5 mM:
Milford, MA) NH4OA-c:Et3N(23:57:20:0.2 by
volume), C: MeOH: MeCN:5 mM:
NH4OAc:Et3N(26:64:10:0.2 by
volume)

Journal of Ethnopharmacology 284 (2022) 114647


Vinblastine HypersilBDS-C18 5 mM Na2HPO4 x 2H2O buffer (pH UV218 Gradient 1.5 Leaves alkaloidal Verma et al. (2007)
(150 × 4.6 mm, 5 adjusted to 7.3 by H3PO4)-
μm) (Agilent) acetonitrile-methanol
Ajmalicine, Bondpak C18 Acetonitrile and ammonium UV254 Gradient 20.52–41.42 Leaves, Root- Ataei-Azimi et al.
cataranthine, (1500 × 2.5 mm, carbonate buffer pH 7.3 (1:1) callus and (2008)
vinblastine, 4.6 μm) roots
Serpentine,
vincristine and
vindoline
Catharanthine and Nucleosil 5 C18 50 mM sodium phosphate pH 3.9: UV280 Leaves Dichloromethane Ramani and
vindoline (250 × 4.6 mm, 5 acetonitrile: 2 methoxyethanol Suspension Jayabaskaran
μm) (Novapak) (80:15:5 v/v)] cultures (2008)
Ajamalicine, 0.01M phosphate buffer (pH 4.9) UV220 Gradient C. Roseus/ 90% ethanolic Singh et al. (2008)
catharanthine, and acetonitrile root, stem,
(continued on next page)
S. Kumar et al.
Table 4 (continued )
S.N. Instrument Analytes Column Mobile Phase Detection Elution Range of RT Flow Plant part Extract Reference
program (min) rate
(mL/
min)

serpentine, Phenomenex C18 leaf, flower


vincristine, (250 × 4.6 mm, and seed
vinblastine and 5¼ m)
vindoline
Catharanthine, Phenomenex 10 mM ammonium acetate buffer UV220 Gradient 0.3 Leaves methanolic Verma et al. (2008)
vinblastine, Gemini C18 (150 (pH 5.0), acetonitrile and methanol
vincristine and × 2.0 mm, 5 μm)
vindoline (Torrance, CA,
USA)
Vincristine Merck LiChro Acetonitrile: methanol; 3:1 UV220 Isocratic 1 Leaf, root and alkaloidal Aslam et al. (2009)
CART C18 (125 × whole Plant)
4.6 mm, 5 μm)
Vincristine Supelco ODS Methanol:Water (50:50 v/v) UV280 Isocratic 0.5 Leaf, stem Methanolic Kalidass et al.
Hypersil, C18 (4.6 (2009)
× 250 mm, 5 μm)
Vinblastine and Nova pack C18 Phosphate buffer solution of pH 6.5, UV Isocratic 8.05–11.83 6 C. Roseus Methanolic, Shams et al. (2009)
vincristine (3.9 × 150 mm) acetonitril (55 : 45) Alkaloidal
(Waters)
А-3′ ,4′ - Waters ODS C18 A: water∶ diethylamine, pH to 7.2 UV220 Gradient – 1.5 Leaves Methanolic Tang et al. (2009)
anhydrovinblastine, (250 × 94.6 mm, H3PO4, B: methanol∶ acetonitrile =
catharanthine and 5 μm) 4∶1
vinblastine, vindoline
Vinblastine Microsorb-MV Methanol and acetonitrile UV240 Gradient 7.02 0.2 Leaves Aqueous Ahmed et al.
C18 (150 × 4.6 (2010b)
24

mm, 5 μm)
Vinblastine Merck LiChro Acetonitrile and methanol, 3:1 UV255 – 1 Leaves/leaf Methanolic Aslam et al. (2010)
CART C18 (125 × callus and in
4 mm, 5 μm) germinating
embryos
Catharanthine, ODS C18 (250 × 4 Water (containing 0.005mol.L-1 UV280 Gradient 7.52–31.36 1 Leaves alkaloidal Tang and Rao
tryptamine and mm) Agilen ammonium hydrogen phosphate), (2010)
vindolin Hypersil, methanol (containing 0.67%
California, USA) triethylamine), and acetonitrile
Ajmalicine, Phenomenex 50% Methanol/50% Acetonitrile: 5 UV218 – 1 Hairy root Methanolic Li et al. (2011)
catharanthine, Luna 5A C18 (250 mM (NH4)2HPO4 (pH 7.3) cultures
lochnericine, × 4.6 mm)
serpentine and (Torrance, CA)
tabersonine

Journal of Ethnopharmacology 284 (2022) 114647


Catharanthine, Zorbax Eclipse Methanol:acetonitrile:ammonium UV Isocratic 10.26–19.62 1 Leaves, stem, Methanol and Siddiqui et al.
vinblastine, vindoline plus C18 (250 × acetate buffer (25 mM) with 0.1% and flower methanol: water (2011)
and vincristine 4.6 mm, 5 μm) triethylamine (15:45:40 v/v)
Catharanthine, HiQ sil-C18 (4.6 × Methanol: acetonitrile (4:1, v/v, UV220 – 10.0–26.0 1.5 Leaves 85% ethanolic Yang et al. (2011)
vinblastine and 250 mm, 5 μm) solution A) water:diethylamine
vindoline, (KYA TECH) (986:14, v/v) with phosphoric acid
pH of 7.5 (solution B).
Catharanthine, HiQ sil C18 (4.6 × A (methanol/acetonitrile), and UV220 Gradient 9.0, 11.9, 2 Leaves 80% ethanolic Mu et al. (2012)
vinblastine, 250 mm) solution B (water/diethylamine, pH 13.2, 20.2
vincristine and 7.2 by H3PO4)
vindoline
β-carotene C18 (250 × 4 mm, Methanol (A), water/methanol UV240 Gradient 0.5 Cell Acetone Thabet et al. (2012)
3 μm) (20:80, v/v) containing 0.2% suspensions
leaves
(continued on next page)
S. Kumar et al.
Table 4 (continued )
S.N. Instrument Analytes Column Mobile Phase Detection Elution Range of RT Flow Plant part Extract Reference
program (min) rate
(mL/
min)

Langerwehe, ammonium acetate (B) and


Germany) tertmethyl butyl ether (C)
Ajmalicine, C18 (Acquity, 98.9% Deionised water, 1.0% UV Isocratic 1 Leaves Homogenised Andrade et al.
catharanthine, Waters, acetonitrile and 0.1% formic acid with acid (2013)
serpentine, Manchester, UK) water+0.01%
vinblastine vindoline formic acid) 1:1
and vincristine (m/v)
Ajmalicine, Mediterranean Methanol-acetonitrile-ammonium MS Gradient 1 leaves Alkaloidal Fernández-Pérez
catharanthine, sea C18 (150 × acetate (0.0025 mol/L) and et al. (2013)
lochnericine and 4.60 mm, 3 μm triethylamine (0.0072 mol/L)
tabersonine Teknokroma, (10:40:50:0.1)
Spain
Vinblastine and C18 (Waters) 5%–95% Acetonitrile in water with UV254,220 Gradient 0.5 Endophytic Alkaloidal Kumar et al. (2013)
vincristine 0.01% trifluoroacetic acid fungus
Fusarium
oxysporum
Catharanthine, himpak VP- Methanol, acetonitrile, and 5 mm UV220 Isocratic 1 Suspension Methanolic Pandiangan et al.
tryptamine, ODSC18 (150 × diammonium hydrogen phosphate culture (2013)
vinblastine and 4.6 mm) (3:4:3)
vindoline
Ajmalicine C18 (4.6 × 150 UV220 Gradient Root Methanolic Thakore et al.
mm 5 μm) (2013)
(Agilent
Technologies)
25

Tryptophan C18 (100 × 4.6 21:79 (v/v) acetonitrile: 0.1 M UV254 Gradient 1.2 Cell alkaloidal Verma et al. (2013)
mm) phosphate + 0.5% glacial acetic suspensions
acid of
Catharanthine and Capcell pak C18 Acetonitrile 20 mM sodium- UV250 Gradient 1 Leaves Methanolic Fukuyama et al.
vindoline (Shiseido Co., phosphate buffer (pH 7.0) (2015)
Ltd., Japan) acetonitrile
Anhydravinblastine, Kromasil C18 Water: diethylamine (986:14, v/v) UV220 2 Leaves 80% Ethanolic Luo et al. (2014)
catharanthine, (250 × 4.6 mm, 5 with phosphoric acid to a pH of 7.5
vinblastine, μm) (solution A) and methanol:
vincristine and acetonitrile (4:1,v/v, solution B).
vindoline
Vinblastine and Octadecylsilane Acetonitrile: Water 50:50 UV Isocratic 1.229–1.474 2 Leaves Methanolic Saravanan and
vincristine C18 (250 × 4.6 Karthi (2014)
mm, 5 μ (Supelco)

Journal of Ethnopharmacology 284 (2022) 114647


Ajmalicine C18 (4.6 × 150 0.01 M phosphate buffer (pH 4.9) UV220, Gradient 11 1.8 Hairy roots alkaloidal Thakore et al.
mm, 5 μm and acetonitrile 254 (2015)
(Agilent
Technologies
1200 series, USA)
Ajmalicine, Phenomenex Methanol/acetonitrile/10 mM UV280 – 1 Suspension- 75% ethanolic Zhou et al. (2015)
catharanthine and Gemini C18 (250 ammonium acetate (15:40:45,v/v/ cultured cells
vindoline × 4.6 mm, 5 μm) v)
(Phenomenex
Inc., Torrance,
CA, USA)
Catharanthine, C18 (250 × 4.6 0.025M ammonium acetate UV220 Gradient 9.00–33.00 1 Leaves Methanolic Zhu et al. (2015)
ajmalicine, mm) Waters solution and acetonitrile
(continued on next page)
S. Kumar et al.
Table 4 (continued )
S.N. Instrument Analytes Column Mobile Phase Detection Elution Range of RT Flow Plant part Extract Reference
program (min) rate
(mL/
min)

strictosidine and
vindoline
Ajmalicine and C18 (150 × 2.1 A: 0.1% formic acid in water, B: UV254 Gradient 12.34–13.09 0.2 Hairy root Methanolic Benyammi et al.
catharanthine mm, 5 μm) 0.1% formic acid in methanol MS lines (2016)
Alltima, Alltech,
Saint Jean de
Gonville, France)
Vinblastine, C18 (250 × 4 mm, 55% sodium phosphate buffer (5 UV Gradient NA 0.7 Root Methanolic Hanafy et al. (2016)
vincristine and 5 μm) mmol/l, pH 6), 45% acetonitrile)
catharanthine
Vindoline C18 (250 × 4.6 70:30 (v/v) mixture of 100 mM MS Gradient 1 Leaves tissues Methanolic Pandey et al. (2016)
mm, 5 μm) ammonium acetate (pH 7.3) (A):
(Waters) acetonitrile (B)
Vinblastine Agilent Zorbax Ammonium acetate (20 mM) 70:30 UV254 Gradient 0.2 Leaves Carbon dioxide Falcão et al. (2017)
Eclipse XDB-C18 acetonitrile, at 30 and ethanol
Ajmalicine, Diamonsil C18 water: diethylamide(v/v), pH 7.2], UV220 Gradient NA 1.5 Leaf, root Methanolic Hashemabadi et al.
catharanthine, (250 × 4.6 mm, 5 methanol: acetonitrile = 4:1 (v/v) (2018)
vinblastine, μm)
vincristine and
vindoline
Ajmalicine, C18 (50 × 2.1 mm, Water (solvent A) and acetonitrile UV Gradient 5.47–11.25 0.4 Root, stem, 70% Methanolic Jeong and Lim
catharanthine, 1.7 μm) (solvent B) containing 0.1% formic and leaf (2018)
serpentine, acid ammonium formate
vinblastine,
26

vincristine and
vindoline
Catharanthine, Eclipse XDB-C18 A and TFA-water-acetonitrile UV254 Gradient 1 CMC Methanolic Moon et al. (2018)
vinblastine, (4.6 × 250 mm, 5 (0.01:5:95) as solvent B suspension
vincristine and μm) (Agilent culture
vindoline Technologies)
Vinblastine Microsorb - MV Methanol – phosphate buffer (5 UV254 Gradient 20 2 Leaves Aqueous Rahim et al. (2018)
C18 (250 × 4.6 mM, pH 6.0) – acetonitrile
mm, 5 μm)
Vincristine Zorbax SB-C18 Methanol-15 mmol⋅L− 1 UV254 1 Aerial parts Carbon dioxide Karimi and Raofie
(2.1 × 50 mm, ammonium acetate containing and ethanol (2019)
1.8 μm) 0.02% formic acid (65: 35, V/V)
Kaempferol, Athena C18 (250 Acetonitrile, Phosphate buffer (pH UV254 Gradient 2.42–9.46 1 Leaves Ethanolic Rao and Ahmed
quercetin and rutin × 4.6 mm) = 5.8) powders (2014)

Journal of Ethnopharmacology 284 (2022) 114647


Gallic acid, quercetin hypersil gold C18 Acetonitrile, methanol, and 3% UV254 Gradient Root, stem, Deep eutectic Nisar (2017a);
and rutin (250 × 4.6 mm, 5 formic acid (50/50/0.3, v/v/v) leaves, and solvent Nisar (2017b)
μ) flower petal
7,3-O- Phenomenex 3% Aqueous formic acid (A) and UV520 Gradient 0–40 min 1 Flower petals Natural deep Dai et al. (2016)
dimethylcyanidin, 3- Luna C18 (4.6 × methanol with 3% formic acid eutectic solvents
O-(6-O-p-coumaroyl) 250 mm, 5 μm)
glucose, hirsutidin 3-
O-(6-O-p-coumaroyl)
glucose, 7-O-
methylcyanidin,
malvidin 3-O-(6-O-p-
coumaroyl) glucose
and petunidin
2. UPLC UV280 Isocratic 1 Leaves 75% ethanolic Zhou et al. (2015)
(continued on next page)
S. Kumar et al.
Table 4 (continued )
S.N. Instrument Analytes Column Mobile Phase Detection Elution Range of RT Flow Plant part Extract Reference
program (min) rate
(mL/
min)

Ajmalicine, Phenomenex Methanol/acetonitrile/10 mM


catharanthine and Gemini C18 (250 ammonium acetate (15:40:45, v/v/
vindoline × 4.6 mm, 5 μm) v)
3. HPLC-MS Catharanthine, Hypersil ODS Solvent A: 25 mM CH3COONH4 in MS Gradient 14.8–21.3 1 Leaf Isopropyl Chu et al. (1997)
vinblastine, (200 × 4.6 mm, 5 methanol CH3COONH4 in water; alcoholic
vincristine, μm) (Agilent) Solvent B: 25 mM
vindoline,
vindolidine,
vindolinine and 19S-
vindolinine
Vinblastine and Zorbax Bonus C18 Solvent A: 20 mM CH3COONH4; MS Gradient 18–21.5 0.2 Aerial parts Methanolic Choi et al. (2002)
vincristine (150 × 2.1 mm, 5 Solvent B: Acetonitrile and roots
μm) (Agilent)
Vinblastine, Direct flow Methanol MS N/A – Whole plant Commercial Zhou et al. (2005)
vincristine, vindoline injection
and vindolidine
Vinblastine and Xterra MS C18 10 mM ammonium acetate (pH 10) MS Isocratic 1 Leaves Ethanolic Rischer et al. (2006)
vincristine (4.6 × 150 mm, 5 and acetonitrile (55:45) Suspension
μm) (Waters) cultures
Vincristine, Phenomenex Solvent A (46%) + 0.1% TFA/ MS Isocratic 5–14.40 0.2 Plasma Schmidt et al.
vinblastine and Polar-RP (250 × water; solvent B (54%) + 1:1 (2006)
vinorelbine 2.0 mm, 4 μm) methanol: acetonitrile.
Ajmalicine, Luna C18 (250 × acetonitrile (A) and acetic acid 1% MS Gradient 10.0–40.0 1 Root Methanol: water Ferreres et al.
ajmalicine (isomer), 4.6 mm, 5 μm) (B) (1:1) (2010)
27

catharanthine, (Phenomenex,
serpentine, United Kingdom)
serpentine (isomer),
tabersonine,
vindolinine and 19-S-
vindolinine
Vinblastine Merck LiChro Acetonitrile: methanol (3:1) UV255 Gradient 1 Embryos/ Chloroform Aslam et al. (2011)
CART C18 (125 × tissues
94 mm, 5 μm)
Ajmalicine, Waters Acquity Acetonitrile (A) and 0.1% formic MS Gradient 2.08–3.03 0.3 Cell line 80% ethanolic He et al. (2011)
catharanthine, UPLC®BEH C18 acid in water (B). c20hi
serpentine and (50 × 2.1 mm,
vindoline 1.7 μm)
Vindoline Acquity Ultra 50∶50 Acetonitrile-formic acid in MS Isocratic 0.5 Leaves Methanolic Liscombe &
BEH C18 (100 × water alkaloidal O’Connor (2011)

Journal of Ethnopharmacology 284 (2022) 114647


2.1 mm, 1.7 μm)
Ajmalicine, C18 (Acquity, 98.9% Deionised water, 1.0% MS Isocratic 1 Leaves Homogenised Andrade et al.
catharanthine, Waters, acetonitrile and 0.1% formic acid with acid (2013)
serpentine, Manchester, UK) water+0.01%
vinblastine, formic acid) 1:1
vincristine and (m/v)
vindoline
Ajmalicine, DL-Cl8 (250 × 4.6 Acetonitrile (A) and 10 mmoL UV, MS Gradient 1–0.5 Leave Alkaloidal Chen et al. (2013)
catharanthine, mm, 5.0 μm) ammonium acetate in water (B)
vinblastine vindoline (Japan)
and vinleurosine
Catharanthine; BEH C18 (50 mm 0.1% formic acid (A) and MS Gradient 1.74–4.47, 0.5 Leaf Methanol: water El-Domyati et al.
serpentine, × 2.1 mm, 1.7 acetonitrile (B) (1:1) (2014)
taberosinine, μm)
(continued on next page)
S. Kumar et al.
Table 4 (continued )
S.N. Instrument Analytes Column Mobile Phase Detection Elution Range of RT Flow Plant part Extract Reference
program (min) rate
(mL/
min)

vinblastine,
vincristine,
vindoline,
vindolidine,
vindolinine and 19-S-
vindolinine
Catharanthine, Agilent Eclipse Methanol mmol/L ammonium MS Gradient 2.51–2.97 1 Leaves, stems, Water− 0.1% Zhang et al. (2014)
vinblastine, C18 (150 × 4.6 acetate containing 0.02% formic (MRM) roots H2SO4 methanol
vincristine and mm, 5 μm) acid (65 : 35, V/V)
vindoline,
vinleurosine
Catharanthine and C18 (2.1 × 50 mm, Acetonitrile–0.1% formic acid in MS Gradient 0.4 Plasma Lin et al. (2015)
vindoline 3.5 μm) water
Ajmalicine, Xselect CSH solvent A: water with 0.1% formic MS Gradient 7.0–13.0 0.3 Roots 80% methanolic Nakabayashi et al.
catharanthine, Phenyl-Hexyl acid) and solvent B: (acetonitrile (2015)
hirsuteine, hirsutine, (2.1 × 150 mm, with 0.1% formic acid)
formosanine, 3.5 μm) Waters,
isoformosanine, Milford, MA,
isorhynchophylline, USA)
isopteropodine,
mitraphylline
pteropodine and
rhynchophylline
Catharanthine, Phenomenex methanol/acetonitrile/10 mM MS Isocratic 1 Root-callus 75% ethanolic Zhang et al. (2015)
28

vinblastine, Gemini C18 (250 ammonium acetate (15:40:45, v/v/ and roots
Vincristine and × 4.6 mm, 5 μm) v)
vindoline,
Catharanthine, Cosmosil C18 (4.6 (A) 0.05 mol/L ammonium acetate MS Gradient 4.88–36.37 1 Leaves and Water: methanol Liu et al. (2016)
serpentine, × 250 mm, 5 μm), and (B) acetonitrile, Acetonitrile/ Human serum (50:50, v/v) and
tabersonine, WondaSil C18 0.2% formic acid in water, 37:63, v/ samples alkaloidal
tryptamine, (200 × 4.6 mm, 5 v
vinblastine, μm)
vincamine and
vindoline and
yohimbine
16 alkaloids Eclipse XDB-C18 25 mM ammonium acetate and LTQ Isocratic 0.25 Stem tissue Yamamoto et al.
(150 × 4.6 mm, 5 acetonitrile Orbitrap at 20:80 (2016)
μm) (Agilent)
Vinblastine Zorbax XDB C18 Methanol and Nano pure water with MS Isocratic 4.91–5.2 1 Leaves 90% Ethanolic Ayob et al. (2017)

Journal of Ethnopharmacology 284 (2022) 114647


(150 × 4 mm, 5 0.01% acetic acid
mμ)
Ajmalicine, VP-ODS C18 (200 (A) 0.04% HoAc− H2O, (B) 0.04% MS – 0.5 Leaves 70% methanolic Zhang et al. (2017)
serpentine, × 150 mm, 5.0 HoAc− MeCN
vinblastine and μm)
vincristine
22 alkaloids C18 (2.1 × 50 mm, Water (solvent A) and acetonitrile MS Gradient 1.8–13.4 0.4 Root, stem, 70% Methanolic Jeong and Lim
1.7 μm) (solvent B) containing 0.1% formic and leaf (2018)
acid ammonium formate
61 alkaloids Thermo Вsil C8 using 0.1% formic acid in water and MS Gradient 1–37 min 0.5 Leaf, stem Ethanolic Kumar et al.
(250 × 4.5 mm, 5 acetonitrile and root) (2018a)
μm)
0.1% aqueous FA and acet-onitrile HPLC-MS Gradient 0.6 leaves Choline chloride, Koel et al. (2020)
(containing 0.1% FA) Menthol
(continued on next page)
S. Kumar et al.
Table 4 (continued )
S.N. Instrument Analytes Column Mobile Phase Detection Elution Range of RT Flow Plant part Extract Reference
program (min) rate
(mL/
min)

vincamine, Eclipse Plus C18


catharanthine and (150 × 2.1 mm,
vindoline 3.5 μm)
18 phenolics LiChroCART C18 1% Acetic acid (A) and acetonitrile UV, MS Gradient 1.0–30.0 1 Stems, leaves Aqueous Ferreres and Pereira
(250 × 4 mm, 5 (B) and petals (2008)
μm) (Merck,
Darmstadt,
Germany)
7,3-O- Kinetex C18 (A) 0.1% formic acid (v/v) in water; LC–MS Gradient 0–20.5 0.3 Flower petals Natural deep Dai et al. (2016)
dimethylcyanidin, (B) 0.1% (v/v) formic acid in eutectic solvents
hirsutidin 3-O-(6-O- methanol
p-coumaroyl)
glucose, 7-O-
methylcyanidin,
malvidin 3-O-(6-O-p-
coumaroyl) glucose
and petunidin 3-O-(6-
O-p-coumaroyl)
glucose
29

4. UPLC-MS Vindogentianine Waters XBridge A (0.1% formic acid in water) and B MS Isocratic – 1.5 Leaves Dichloromethane Tiong et al. (2015)
C18 (50 × 2.1 mm, (0.1% formic acid in MeOH) and methanol
2.5 μm)
5. UHPLC– Ajmaline, ajmalicine, ACQUITY UPLC acetonitrile, methanol and formic MS Gradient 2.09–5.92 0.5 Leaf, stem Ethanolic Kumar et al.
MS serpentine, reserpine, BEHTM C18 (2.1 acid root) (2018b)
vinblastine, × 50 mm, 1.7 μm)
vincristine,
vindesine, vindoline,
vinpocetine and
yohimbine
6. CE-MS Catharanthine, (6500 × 50 μm) 20 mM ammonium acetate and MS Directly 13–17 0.4 Stem Methanolic Chen et al. (2011)
vinblastine and (Yongnian, Hebei, 1.5% acetic acid injected
vindoline China)

Journal of Ethnopharmacology 284 (2022) 114647


S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Table 5
Pharmacological activities and key results of different extract of plant parts of C. roseus.
S. Pharmacological Compounds/plant Extract/Class of compound Dose Result Reference
N. activity part

Anticancer and Leaves Ajmalicine, vindorosine, – Ajmalicine and vindorosine reports for the Tin-Wa et al. (1968)
cytotoxic lochnerinine cytotoxic alkaloid
1. Leaves Aqueous 2.55, 2.38 μg/ The crude aqueous extract of C. roseus shows Ahmed et al. (2010b)
mL differential effects of inhibiting the proliferation
in different cell lines
Leaves Catharanthine 60 μg/mL Catharanthine showed the most considerable Siddiqui et al. (2010)
activity
Stem Methanol, n-butanol, aqueous 5.2–21.0 μg/mL n-butanol fraction prepared from C. roseus stem Pham et al. (2018)
fraction antitumor therapeutic agents.
suspension Indole alkaloid-enriched 211, 210 ng/mL Indole alkaloid-enriched bioactive extract is Fernández-Pérez
cultured-cells bioactive extract powerful antitumor activity et al. (2013)
Whole plants Dimeric indole alkaloids – All these compounds were evaluated for their in Wang et al. (2012b)
vitro cytotoxic activities against human breast
cancer cell line.
– 50 alkaloids 0.1–1 μg/mL All alkaloid fractions showed a dose-dependent Khanavi et al. (2010)
cytotoxic effect on the different cell lines.
– Vinblastine, vincristine – Vinblastine, vincristine showed ctotoxic effects. Ruszkowska et al.
(2003)
2. Antidiabetic Leaves and twigs Dichloromethane:methanol 500 mg/kg Results showed increased metabolization of Singh et al. (2001)
extract (1:1) glucose in treated rats.
Leaves Leaf juice or water decoction 0.5, 0.75, 1.0 The leaf juice showed significant antidiabetic Nammi et al. (2003)
mL/kg BW activity.
Leaves Ethanol 150, 300 mg/kg C. roseus showed lowering blood glucose level. Akhtar et al. (2007)
Leaves Aqueous extract 100 & 500 mg/ The aqueous extract showed significant Mostofa et al. (2007)
kg antidiabetic activity
Leaves Ethanol 30 mg/155 ± 15 C. roseus leaves extract treated animals have Ara et al. (2009)
shown the hypolipidemic effects.
Leaves Petroleum ether, chloroform, 150 mg/kg Chloroform fractions of C. roseus showed high Islam et al. (2009b)
ethyl acetate fractions antiyperglycemic activity
Leaves Aqueous 3, 500 mg/BW 500 mg dose showed significant effects. Prasad et al. (2009)
Leaves Petroleum-ether, ethyl 150 mg/kg Ethyl acetate fractions were the best in activity. Islam et al. (2009a)
acetate, chloroform fractions
Leaves Methanol, n-hexane, 200 μg/mL Chloroform fraction showed the highest activity Siddiqui et al. (2010)
chloroform, water
Leaves Dichloromethane: methanol 500 mg/BW Leaf dichloromethane methanol extract was Jayanthi et al. (2010)
found to exhibit a significant anti hyperglycemic
activity.
Leaves Powder 55 mg/kg BW Potential hypolipidemic properties reported in Rasineni et al. (2010)
leaf extract.
Leaves Powder 100 mg/kg BW C. roseus with its antidiabetic properties could Rasineni et al. (2010)
be a potential herbal medicine in treating
diabetes.
Leaves Aqueous extract 0.30, 1 mL/kg The study showed significant reduction glucose. Ahmed et al. (2010b)
BW
Leaves – 100 mg/kg BW C. roseus showed potent hypolipidemic Rasineni et al. (2010)
properties
Leaves Ethanolic 200, 500 mg/kg Ethanol extract of C. roseus possesses significant Hassan et al. (2011)
antidiarrheal effect.
Leaves – 150 mg/kg BW C. roseus leaves have potent antihyperglycemic Manoharan et al.
effect in alloxan-induced diabetic rats. (2011)
Leaves Methanol 250 mg/kg The leaf extract of C. roseus significantly Ohadoma and
increases the hypoglycemic effect. Michael (2011)
Leaves Leaf juice 0.5, 1 mL/kg Leaf juice of C. roseus possesses significant lipid Patel et al. (2011)
lowering and anti-atherosclerotic activity
Leaves Aqueous 250 mg/Kg The best hypoglycemic activity was found in Vega-Ávila et al.
aqueous extracts. (2012)
Leaves Total phenolic contents – The highest inhibitory effect of 63% is shown by Aadil et al. (2012)
phenolic extract.
Leaves Ethanol 20 mg/kg Ethanolic extract C. roseus exhibited Ahmed and Rao
hepatoprotective activity (2013)
Leaves Vindoline, vindolidine, 25.0 μg/mL Vindolicine induced relatively high glucose Tiong et al. (2013)
vindolicine and vindolinine uptake.
Leaves Aqueous 10 mg/100 g Aqueous extract was found to be effective as an Muralidharan (2014)
antidiabetic agent.
Leaves – – C. roseus fresh leaves showed hypoglycemic Bisla et al. (2014)
activity.
Leaves Aqueous 150 mg/kg Leaves were inactive for hypoglycaemia effect Muralidharan (2014)
Leaves Methanolic 200, 400 mg/kg The methanolic extract of C. roseus significantly Singh et al. (2014)
BW increases the wound contraction in mice that
promises to overcome the delayed wound
healing in diabetic condition.
Leaves Ethanolic 100, 200, Antidiabetic effect of C. roseus on STZ induced Al-Shaqha et al.
diabetes. (2015)
(continued on next page)

30
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Table 5 (continued )
S. Pharmacological Compounds/plant Extract/Class of compound Dose Result Reference
N. activity part

Leaves Ethanol 150 mg/kg Ethanolic extract of C. roseus leaves exhibit Mohan et al. (2015)
antidiabetic activity
Leaves Vindogentianine 200 μg/mL In vitro α-amylase and α-glucosidase inhibition Tiong et al. (2015)
assay showed low inhibition under treatment of
vindogentianine
Leaves Aqueous – Leaf extracts of C. roseus has hipolipidemic Suarsana et al.
activity in hypercholesterolemic rats. (2015)
Leaves Methanol 250 mg/kg BW Methanolic extract of leaf of C. roseus reduce the Aruljothi and
blood glucose level to the rats. Samipillai (2016)
Leaf and flower Ethanol 12.5 mg/mL The alcoholic extract has a main role in Malathi et al. (2010)
carbohydrate metabolism like α-amylase and
α-glucosidase.
Leaves and twigs Total alkaloids extract 100–150 mg/kg The results showed alkaloids extract possessed Zhang et al. (2016)
in antiyperglycemic activity againt STZ induced
hyperglycemic rats.
Leaves, flowers Aqueous 25 mg/mL Aqueous extracts of C. roseus showed Iweala and Okeke
and tender stems considerable hypoglycemic effect. (2005)
Whole plant Methanolic 300, 500 mg/kg The methanolic extract at high dose (500 mg/ Ahmed et al. (2010b)
kg) showed significant antidiabetic activity
3. Antimicrobial Crude extracts/ Ethanolic, methanolic, 100 μg/mL Ethanol showed highest activity whereas Ramya (2008)
leaves, stem, root aqueous aqueous extract was inactive
and flower
Callus Aqueous extract of callus – C. roseus callus extract showed significant Batra et al. (2004)
antibacterial effect.
Hairy roots/wild- Methanolic 100–300 μg/mL High antimicrobial activity reported in the Hanafy et al. (2016)
type roots extracts of the transgenic hairy roots
Leaf, nodal callus, Methanolic – Methanolic extracts possess a great strength to Naz et al. (2015)
fruit fight against the microbial activity
Leaves Ethanol – Leaf extract possesses antibacterial activity Khalil (2012)
Leaves Acetone, ethanol,,chloroform 100 mg/mL Acetone, ethanol and chloroform extracts have Shanmugaraju and
inhibitory effect against most of the pathogenic Bhakyaraj (2016)
microorganisms.
Leaves Methanol ethanol acetone – Leaf extracts show significant antimicrobial Patil and Ghosh
activity (2010)
Leaves Ethanol, acetone, aqueous 25–50 μg/μL All extracts showed significant inhibition Kumari and Gupta
against selected microbes. (2013)
Leaves Aqueous, ethanolic/its 5, 10, 20, 40, 80 Highest antifungal activity was found in Roy & Chatterjee
fractions of petroleum ether, mg/mL chloroform fraction (2010)
diethyl ether, chloroform
Leaves Water, ethanol, methanol 25–100 μg/disc Leaf extracts having the potential to inhibit the Balaabirami &
acetone, hexane growth of bacteria and fungi. Patharajan (2012)
Leaves, shoot, and Alkaloids – Antimicrobial activity against Vardhan et al. (2012)
root multiphytopathoges
Leaves, stem, Ethanol and methanol 100–300 μg/mL The ethanolic extract showed potential Govindasamy and
flower and root antibacterial activity. Srinivasan (2012)
Leaves, stem, 95% ethanol The root extract was most effective which Raza et al. (2009)
flower, root exhibited broad-spectrum antibacterial activity
Leaves, stems and Ethanolic, methanolic, – Ethanolic extract showed high antibacterial Chaman et al. (2013)
flowers aqueous, chloroform while methanolic extracts reported for high
antifungal activity.
Leaves, stems, Methanol, acetone, ethyl 250–1000 mg/L Ethyl acetate extracts of different plant parts Sathiya et al. (2008)
roots and flowers acetate were found to possess best antibiogram followed
by methanol and acetone extracts.
Root Hexane, chloroform, ethyl 250–1000 μg/ Chloroform extract was found to be most active Ethalsha and Retna
acetate, methanol, aqueous mL amongst tested extracts (2014)
Seeds – Antibacterial activities were observed in the Kumar et al. (1997)
seeds of C. roseus
Stem Methanol/its fractions n- – n-Butanol fraction showed highest antimicrobial Pham et al. (2018)
butanol, n-hexane, aqueous activity
Water, methanol – Methanol extract was found to be the best for Kabesh et al. (2015)
antibacterial activity.
4. Antioxidant Cell suspensions Crude alkaloid 50 μg/mL This crude alkaloid extract showed strong Verma et al. (2013)
activity
Leaf callus Compound RF1 1.0 mg/mL Compound RF1 was found to possess strong Verma et al. (2012)
cultures antioxidant potential.
Leaf, stem, flower, Water, methanol, ethanol, – Water extract of leaf shows high antioxidant Pham et al. (2018)
and root ethyl acetate activity.
Leaves Methanol, aqueous 50–250 μg/mL It exhibited potent free radical scavenging and Aadil et al. (2012)
antioxidant activity.
Leaves Vindoline, vindolidine, 0–25.0 μg/mL Vindolicine highest antioxidant potential. Tiong et al. (2013)
Vindolicine, vindolinine
Leaves Ethanol 55.2, 26.22, Ethanol extract of C. roseus does not have Widowati et al.
728.05, 404.76 antioxidant activity. (2011)
μg/mL
Leaves and twigs Total alkaloids 150 mg/kg CTA showed strong antioxidant effect Zhang et al. (2015)
(continued on next page)

31
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Table 5 (continued )
S. Pharmacological Compounds/plant Extract/Class of compound Dose Result Reference
N. activity part

Root Hexane, chloroform, ethyl 100–1000 μg/ Crude chloroform extract of C. roseus showed Ethalsha & Retna
acetate, methanol, aqueous. mL high vitro antioxidant activity (2014)
Root and shoot – 0, 50, 100 μM. This plant shows good antioxidant potential. Rai et al. (2014)
Root, leaf, hairy Ethanol 200–400 μg/mL The hairy roots of C. roseus showed considerable Mardani-Nejad et al.
root and callus antioxidant power. (2016)
Roots Ethyl acetate extracts 500–1000 μg/ It shows significant antioxidant activity and Jasmine & Agastian
mL α-glucosidase inhibition (2013)
Roots Aqueous – C. roseus was very good source of natural Pereira et al. (2010)
antioxidants
Seeds, stems, Aqueous extracts 300–1200 μg/ A concentration-dependent protective effect was Ferreres and Pereira
leaves and petals mL observed for seeds and tissues, with petals (2008)
shown to be the most active.
Shoots N–hexane, ethylacetate, 20–100% 100% methanolic extract and 100% ethyl Rasool et al. (2011)
methanol, chloroform acetate fraction of C. roseus shoots exhibit
highest antioxidant activity.
Stem Methanol, n-butanol (b); and – n-Butanol fraction possessed the strongest Pham et al. (2018)
aqueous fraction antioxidant capacity amongst the tested
extracts.
Whole plant Ethanol 125; 62.5; 31.25; C. roseus extract had no antioxidant property. Widowati et al.
15.625; 7.813, (2010)
3.906 μg/mL
5. Larvicidal & Leaves Aqueous, ethyl acetate and 50 mg/mL The aqueous and methanol extracts of C. roseus Subarani et al.
pupicidal methanol leaves had an excellent potential to control the (2013)
malarial vector
Leaves Petroleum ether 20 g/L crude extracts of C. roseus reported to possessed Panneerselvam et al.
the larvicidal potentiality. (2013)
Leaves Aqueous, methanol crude, 1000 ppm All extracts exhibited moderate larvicidal Alaguchamy and
petroleum ether, methanol effects. Jayakumararaj
fraction, ethyl acetate fraction (2015)
Leaves Hexane, ethyl acetate, and – The organic leaf extract of C. roseus showed Pavunraj et al.
methanol significant larvicidal effects. (2017)
Leaves n-hexane fraction of acetone 1 ppm The highest concentration produced 100% Kuppusamy et al.
extract mortality in first to second instars. (2009)
Root Silver nanoparticles 3.5–5.0 mg/mL C. roseus offer potential source for larvicidal Rajagopal et al.
activity against the larvae of both dengue and (2015)
filariasis vectors.

S.N.: Serial Number; BW: body weight; ppm: parts per million.

standard deviation %) and 10.9–10.5 (RE: relative error %). The re­ simultaneous quantitation of MIAs in ethanolic extracts of 39 samples of
covery rate of the alkaloids of samples varies from 79.9 to 91.5%. C. roseus collected from five different locations in India (Kumar et al.,
Inter-day precision and stability of samples showed that the compounds 2018b). Compound 95 was found one of the most abundant compounds
111, 112 and 131 were stable at room temperature, 4 ◦ C and − 20 ◦ C for in C. roseus may be due to conversion of compound 80 to 95 is very rapid
2 h, 12 h and two weeks, respectively. The developed method was as previously reported methods (Kumar et al., 2018b; Jeong and Lim,
applied for quantitation of MIAs and validated with three plant parts of 2018; Liu et al., 2007).
three batches of C. roseus collected from three regions of China (Uniyal
et al., 2001). Similar methodology, the ultra-performance liquid
5.2. Qualitative and quantitative analysis of flavonoids and phenolic
chromatography-quadrupole time-of-flight (UPLC-Q-TOF) mass spec­
acids
trometry method was reported Jeong and Lim (2018) who claimed that
method more sensitive, rapid and novel for quantitation of compounds
Piovan and FilippiniFavretto (1998) and Mustafa and Verpoorte
27, 34, 80, 95, 111 and 112 in extracts of C. roseus. The C18 column has
(2007) have reviewed analytical methods reported for the analysis of
been used for the separation of these alkaloids by mobile phase aceto­
phenolics. Among all ionization sources available, ESI in negative ion
nitrile and water with 0.1% formic acid and 10 mM ammonium acetate
mode is regarded as the most suitable for the analysis of phenolics. The
within 13 min. Compounds 111 and 112 were detected with higher
analysis of phenolics (flavonoids and phenolic acids) was reported by
abundance in the aerial parts whereas the roots showed high content of
both UV–Vis and MS with detection limits for flavonoids in the range of
compounds 27, 34, 80 and 95 (Jeong and Lim, 2018). Similarly, the
10 ng whereas single ion monitoring (SIM) mode provides better
HPLC-ESI-MS/MS method has been reported for the quantitation of
detection limits, usually below 1 ng. One drawback in SIM mode,
selected alkaloids present in trace amounts in C. roseus. The content of
however, causes loss of valuable information concerning fragmentation
compound 131 was reported 2.4–17.6 μg/g, DW (dry weight) in leaf,
patterns, which is very important for the identification and character­
stem, and root of C. roseus samples collected from three different regions
ization of various compounds from plant extracts (Ren et al., 2018;
of China (Zhang et al., 2014). In our findings, sensitive, rapid, validated
Gumbi et al., 2019). Therefore, the phenolic compounds of C. roseus
and reproducible ultra-high-performance liquid chromatography-
have been analyzed using HPLC, HPLC-DAD-ESI-MS, and
electrospray ionization tandem mass spectrometry (UHPLC-E­
HPLC-DAD-ESI-MS/MS (MustafaVerpoorte, 2007; Parthasarathy et al.,
SI-MS/MS) method has been published for simultaneous quantitation of
2020). Piovan and FilippiniFavretto (1998) were studied to understand
compounds 27, 52, 72, yohimbine (77), 78, 80, 95, 111, 112 and 121 in
the distribution of these metabolites within the different plant tissues of
crude extracts of C. roseus in mode using ultra-high-performance liquid
leaves, stems, seeds, and flowers. Fifteen glycosides of isorhamnetin,
chromatography coupled with electrospray ionization hybrid triple
quercetin and kaempferol have been identified and characterized, some
quadrupole-linear ion trap mass spectrometry (UHPLC-E­
of them quantified in extracts of these plants using HPLC and HPLC-MS.
SI-QqQLIT-MS/MS) in MRM mode. The method was applied for the
The synthesis of bioactive compounds depended upon the agricultural

32
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

practices, climate, or life cycle of the plants, therefore compound 162 cytotoxic, antidiabetic, antimicrobial, antimicrobial, antioxidant larvi­
was not detected in some studies reported by Ferreres and Pereira cidal and pupicidal (Gajalakshmi et al., 2013; Nisar et al., 2016; Das and
(2008). Crude extract of roots of C. roseus have been also examined for Sharangi, 2017; Mishra and Verma, 2017; Senbagalakshmi et al., 2017;
the presence of phenolic compounds, but no such phenolics have been Paarakh et al., 2019). Among them the anticancer activity is well known
detected (Ferreres and Pereira, 2008). The two main flavonoids 154 and for this plant. The major pharmacological activities and their method­
150 have been reported in C. roseus leaves and stems (Nishibe et al., ology such as type of extraction, plant parts used, dose and key results
1996; MustafaVerpoorte, 2007). The compound 154 was reported to be which were used during assessment are summarized in Table 5.
abundant in petals, while compound 149 also detected in the petals.
Another isomer with a similar fragmentation was compound 156 6.1. Anticancer activity (cytotoxic activity)
detected only in trace amounts in the stems and leaves (Cui et al., 2000).
Compound 151 has been reported solely in petals in trace amounts. The different extracts of leaves of C. roseus showed significant anti­
Compound 155 has been reported in higher amounts in stems, seeds, cancer activity due to the presence of its bisindole alkaloids (Mishra and
and petals, whereas compound 156 found in seeds of C. roseus. Com­ Verma, 2017; Senbagalakshmi et al., 2017; Paarakh et al., 2019). The
pounds 155 and 156 were detected in very low amounts in seeds, characteristics and major active compounds 112 and 111 of the plants
whereas petals showed an abundant amount of compounds 149, 156, used in chemotherapy for treatment of many types of cancer (Orwa
155, 159 and 160 (Steinmann and Ganzera, 2011; Simirgiotis et al., et al., 2007). In contrast, these compounds isolated from the various
2012). Compound 154 was the most abundant in seeds. Compound 154 extracts of the plants have been reported for cytotoxicity in a wide range
was detected 292.3 mg/kg, DW, and 52.7 mg/kg DW in seeds and leaves, of cell lines. The methanolic extract of C. roseus has been reported for
respectively. The amount of compound 155 was reported in 2.0, 714.2, cytotoxicity against human colorectal carcinoma cell line (HCT 116)
190.8, 8.5, 8, 120.8 mg/kg, DW in seeds, stems, leaves and petals using MTT assay. The highest activity dose-independent cytotoxicity
respectively. Similarly, compound 149 was detected in the amounts reported in chloroform and methanol fractions. The n-hexane fraction
582.7, 190.5, 310.9, 1027.9 mg/kg, DW in seeds, stems, leaves, and showed moderate whereas minor cytotoxic activity occurs in water
petals respectively (MustafaVerpoorte, 2007). fraction (Sagar et al., 2020). Compound 27 has been reported for cyto­
High concentrations of anthocyanins (0.001–0.50 μmol/g fresh toxic activity at dose at 200 μg/mL, whereas the most promising cyto­
weight) were reported in all cells present in a medium induced culture. toxic activity has been observed in compound 55 at the dose-dependent
The compounds 170–172 have been identified and characterized as manner with IC50 value of 60 μg mL (Siddiqui et al., 2010). Three
aglycones of the anthocyanins. The total phenolic content was detected fractions of the alkaloidal extract have been reported for cytotoxic ef­
ranged from 20.0 to 70.0725 mg/g fresh weight) (Knobloch et al., 1982). fects on cell proliferation of HT-29, Caco-2, T47D and NIH/3T3 cell lines
The anthocyanin content in the flowers from both types of plant were due to dose-dependent cytotoxicity. The proliferation of NIH/3T3 cells
comparable and approximately 1.5 times higher than those obtained has been less affected in comparison to other cell lines (Khanavi et al.,
from cell suspensions. Compound 178 was the major compound 2010). The n-butanol fraction have been reported for in vitro cytotoxic
(49.3–70 relatively abundant) in this plant. The extracts from both activities on cell line Du145 (prostate), HT29 (colon), H460 (lung),
embryogenic plant flowers and cell cultures have contained the malvidin A431 (skin), A2780 (ovarian), MIA PaCa-2 (pancreas), MCF-7 (breast
derivatives in a higher amount than the petunidin ones (Filippini et al., SJ-G2, U87), BE2-C (neuroblastoma) and SMA (glioblastoma) at low
2003). Compounds 174, 175, 176, 177, 178 and 179 have been doses (GI50 values of 5.2–21.0 μg/mL) (Pham et al., 2018). The indole
detected at 3.50, 1.50, 11.50, 28.0, 49.3 and 6.20 mg/g, fresh weight alkaloid-enriched bioactive extract with methyl jasmonate (MJ) and
respectively using direct infusion electrospray (ESI) mass spectrometry cyclodextrins (CDs) has been produced cytotoxicity in three cell lines,
and ion trap multiple mass spectrometry in positive-ionization mode. such as THP-1 human monocytic leukemia, Jurkat E.6 human lympho­
The all three 3-O-(6-O-p-coumaroyl) glucosides were identified and cytic leukemia and BL 1395 non-tumor human B-cell line. The cytotoxic
characterized for the first time in C. roseus (Piovan and FilippiniFavretto, assays have been studied using the colorimetric assay 2, 3-bis
1998). (2-methoxy-4-nitro-5-sulfophenyl)-S-[(phenylamino) carbonyl]-2 tetra­
zolium hydroxide (XTT). The Jurkat E.6 and THP-1 cell lines inhibited
5.3. Qualitative and quantitative analysis of volatile compounds by 50% were 211 and 210 ng/mL, respectively. Fernández-Pérez et al.
(2013) claimed that the extract has been exhibited the potent antitumor
The volatile compounds have identified by chromatography-mass activity due to the effect of compounds 111 and 112 but depends on the
spectrometry (GC-MS) in leaves, stems, flowers, and roots of C. roseus. synergistic effect of the compounds 6, 55, 80 and lochnericine (16). The
Mohan et al. (2015) has identified eleven compounds in the ethanolic C. roseus extract has significantly induced the proliferation of PBMCs (P
extract of the leaves and whereas fifteen compounds have been reported < 0.01) taken from normal donors comparing Jurkat cells for cytotoxic
by Doshi et al. (2018) in the same extracts. Seventy six compounds have effects. The maximum cell proliferation was reported in PBMCs incu­
been identified in aqueous extract of the leaves and industrial marcs bated with 1000 μg/mL of the C. roseus extract with increased viability
(Brun et al., 2001). The high of the content volatile compounds has been of 165.93% at 24 h, 117.67% at 48 h and 116.67% at 72 h. The distinct
reported in essential oils of the leaves consisting mainly of linolenic acid effects of inhibition of proliferation of the Jurkat cell line and stimu­
ethyl ester (43.9%) (330), phytol (29.6%) (274), limonene (23.2%) lating the growth of PBMCs due to aqueous extract of C. roseus (Ahmad
(212), stearic acid (10.6%) (314), geraniol (7.3%) (278) and dodecyl et al., 2010a). The cytotoxic activities of eight bisindole alkaloids
alcohol (9.8%) (271) (Lawal et al., 2015). Pandey-Rai et al. (2006) re­ including, catharine (122), leurosidine ( 123), compound 125 and
ported fifty two compounds and accounting showed 91.3% volatile of vinamidine (135) etc have been reported on MDA-MB-231 cells were
the leaf oil. Eighteen compounds have been identified and assessed the tested by MTT assay. These alkaloids might induce a marked reduction
abundances of these compounds in each cultivar. The result showed of the cell viability after 72 h treatment. Among these, the compound
abundance of each cultivar as dark pink (66%), pink (50%), pinkish red 125 has exhibited the most potent inhibitory activity, while compound
(55%), milky white (50%), whitish pink (50%), white (44%), purple 135 has been found the weakest activity. It is found that compound 123
pink (44%) and pale pink (27%) (Fitrianto et al., 2020). showed much weaker activity than that of compound 111. The
17-deacetoxy derivatives of compounds 111, 112, 135, 221 and 222
6. Pharmacological activities have a more potent activity than compound 135, implying the lack of
acetoxy at C-17 seems to be an advantage in increasing cytotoxicity.
Various extracts and isolated compounds of C. roseus has been re­ These results provided clear indication for further structural modifica­
ported a wide range of pharmacological activities such as anticancer and tion in bisindole alkaloids (Rosenthal and Kaufman, 1974; Gomber et al.,

33
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

2010; Wang et al., 2012b; Parthasarathy et al., 2020). (Aadil et al., 2012). Findings also reported that the hepatic and muscle
glycogen content were decreased with C. roseus administration (Rasi­
6.2. Antidiabetic activity neni et al., 2010; Mohan et al., 2015).
Compounds 27 and 38 and vindolicine (140) have been reported
Diabetes is metabolic disorder which are characterized as abnormal better protein tyrosine phosphatase-1B (PTP-1B) inhibition activity
enhancement of the glucose level in endocrine system. Few studies (Tiong et al., 2013); and these also suggest beneficial against
carried out by Muralidharan (2014) and Aruljothi and Samipillai (2016) diabetes-type 2 (Bisla et al., 2014). The hypoglycemic activity in
reported that the application of aqueous extract of C. roseus leaf in methanol leaf extracts using alloxan model for diabetes in rats. The leaf
alloxan-induced diabetic rats has significantly (P < 0.001) decreases the extracts have been significantly increasing the hypoglycemic effect of
blood glucose levels. Finding also reported that total glycerides (TC), metformin (Ohadoma and Michael, 2011; Ahmed et al., 2010b).
low-density lipoprotein (LDL), very low density lipoprotein (VLDL) and Vega-Ávila et al. (2012) reported the blood-glucose-lowering properties
triglyceride (TG) decresease and comes to normal by using C. roseus leaf of organics (hexane, dichloromethane, and methanol) and aqueous
extract. No deaths have been reported in 30 days of the experimental extract in healthy and alloxan-induced (75 mg/kg) diabetic mice, after
period and therefore, it is a good plant species for treatment of diabetes. intraperitoneal administration (250 mg/kg BW. The leaf juice of
In another study, the aqueous herbal extract preparation containing C. roseus has been studied for the hypolipidemic activity in adult guinea
C. roseus at a dose rate of 1 g/kg, A. indica (500 mg/kg), and A. sativum pigs according to pharmacokinetic parameters. The leaf juice at dose of
(1 g/kg) BW has been administered in rats orally for 14 days, and finding 0.5 mL/kg reduced the serum lipid parameters. The high-fat diet de­
suggested that the extract showed significant reduction of blood glucose creases the serum lipid parameters in the tissue-induced leaf juice in the
as compared to patent drug (Mostofa et al., 2007). Similarly, the dose of 0.5 mL/kg (Patel et al., 2011).
comparative study of some plant species of other genus have been re­
ported using the aqueous leaf extract of M. koenigii, P. guajava and 6.3. Antifertility effect
C. roseus at dose of 500 mg/kg treated in streptozotocin-induced diabetic
rats (SIDRs) was carried out by different co-workers. They are reported Bioefficacy of ethanol leaf extract of three plant species of C. roseus,
significant body weight gain and blood glucose level in the Ocimum sanctum, and Lantana camara were tested for fecundity and
streptozotocin-induced diabetic rats (SIDRs) after fifteen days of treat­ fertility of the red cotton bug, Dysdercus koenigii. The oviposition
ment (M. koenigii and P. guajava: (P < 0.05), C. roseus, and GBC: (P < behavior, fecundity and fertility of the females who emerged from the
0.001). Findings suggested the aqueous leaves extract of M. koenigii, treated nymphs were assessed. D. koenigii exhibited a reduction in their
P. guajava and C. roseus possessed beneficial effects in diabetic mani­ reproductive success in response to the plant extracts. The ethanol
festations (Prasad et al., 2009; Iweala and Okeke, 2005; Zhang et al., extract of C. roseus was most effective in the suppression of oviposition
2016). Allied to these studies, Islam et al. (2009a) reported for fractions among the examined above plants extracts. The fertile mated females lay
of petroleum-ether and ethyl acetate significantly reduced blood glucose eggs, indicating the insemination of the mated females. However, the
levels in normoglycemic rats (RGRs) and SIDRs. The antidiabetic and percentage of hatchability of the eggs laid by the females was less. The
hypolipidemic activities have been compared to metformin HCl at the eggs laid by the females treated with ethanol extract of Osmium leaf was
dose of 150 mg/kg. The C. roseus fractions in ethyl acetate found to be least fertile. It was suggested that some of the compounds present in the
the best for antidiabetic functions (Islam et al., 2009a, 2009b). extracts individually or synergistically alter the fecundity and fertility of
Nammi et al. (2003) have been studied the leaf juice of C. roseus and D. koenigii (Kayesth et al., 2017). Similarly, petroleum ether extract of
treated it to normal and diabetic rabbits, and also findings were the leaves of C. roseus inhibited the estrogen-induced gain in the uterine
compared with the standard drug and glibenclamide. They reported the weight when administered along with estradiol (Gupta, 2009). The
reduction of glucose level after treatment by juice in a dose-dependent long-term use of the leaves of C. roseus was caused by a reduction in
reduction in blood glucose of the both normal and diabetic rabbits. epididymal sperm count and lowering of serum testosterone. The
The alcoholic flower and leaf extract was tested for alpha-amylase and aqueous leaf extract was orally administered to the animals daily for 30
alpha-glucosidase inhibitory activity and found that the leaf extracts has days. After 30 days of treatment with extract significantly reduced the
the maximum inhibitory action at a dose of 10 mg/mL (IC50). These epididymal sperm count and serum testosterone level respectively
concluded that the leaves and flowers of C. roseus have been exhibited (Sukumar and Osmani, 1981; Shaw et al., 2017).
the anti-diabetic effect by lowering α-amylase and α-glucosidase
(Malathi et al., 2010). 6.4. Antimicrobilal activity
The methanolic fractions of leaf at a concentration of 200 and 400
mg/kg BW (body weight) has been optimal and induced through The crude extracts of various plant parts of C. roseus have been re­
intraperitoneal in mice (Ahmed et al., 2010). The methanolic extract ported for antibacterial activity against several bacterial strains by disc
was significantly increases the contraction of wounds and the formation diffusion assay. Among the different plant parts tested, the leaf extract
of collagen fibers in the mice compared with controls (P < 0.001). A exhibited significantly higher efficacy (Gajalakshmi et al., 2013; Nisar
methanolic fraction of fresh leaves has been treated to rats induced with et al., 2016; Das and Sharangi, 2017; Mishra and Verma, 2017; Senba­
alloxan to decrease the glucose level of blood and found that the serum galakshmi et al., 2017; Paarakh et al., 2019). Similarly, organic solvent
protein recovered the normal glucose level in rats (Singh et al., 2014). extracts showed higher antibacterial activity in comparison with their
Al-Shaqha et al. (2015) have been reported the antidiabetic efficacy of corresponding aqueous extracts (Gajalakshmi et al., 2013; Nisar et al.,
the ethanolic extract and expressions of GLUT-2 and GLUT-4 gene in 2016). Aqueous extracts of C. roseus generally were inactive against
SIDRs. The doses administered orally at 100 and 200 mg/kg, destroys selected bacterial strains. Ethanol extracts found to have good activity
the glucose carrying system of liver within twenty-eight days. A com­ against the tested bacterial strains. The gram negative (-ve) stains were
bination of dichloromethane (DCM) with methanol extract of leaves and found to have more responsive in comparison to gram (+ve) strains of
twigs in ratio of 1:1 at a concentration of 500 mg/kg given orally for bacteria (Ramya, 2008; Hanafy et al., 2016; Fitrianto et al., 2020). Be­
seven and fifteen days which have been shown 48.6 and 57.6% hypo­ sides, the alcoholic extracts (ethanol and methanol) have been tested
glycemic activity using SIDRs model (Singh et al., 2001; Jayanthi et al., against various human pathogens such as Bacillus subtilis, E. coli,
2010). The methanolic and aqueous leaf extracts of Acacia arabica and Staphylococcus aureus, Salmonella typhi, and Pseudomonas aeruginosa. The
Murraya koeingii, C. roseus and Rauvolfia serpentina have been studied for ethanolic and methanolic of found to have the minimum zone of inhi­
α-amylase inhibition. The maximum α-amylase inhibition and glucose bition (MZI) as 21.15 ± 1.64 mm and 15.61 ± 1.35 mm, respectively,
diffusion rate were found in methanolic extracts of Rauvolfia serpentina against the bacterial strain of S. typhi, whereas the MZI observed against

34
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

S. aureus was 06.24 ± 0.69 mm in the fraction of ethanolic extract and extracts have been checked against P. aeruginosa, S. typhimurium, and
MZI observed in methanolic fraction was 05.20 ± 0.86 mm against S. aureus. All these extracts showed antimicrobial activity against
E. coli (Srinivasan et al., 2001; Govindasamy and Srinivasan, 2012). selected microorganisms (Patil and Ghosh, 2010; Kabesh et al., 2015).
The agar-well diffusion method was employed to test the antimi­ The C. roseus n-butanol extract/fraction was observed effectively
crobial activity using the ethanolic, methanolic, aqueous and chloro­ inhibiting the activity of E. coli and Staphylococcus lugdunensis (Pham
form fractions/extracts of different plant parts (flowers, stems, leaves, et al., 2018).
roots) of C. roseus against E. coli, P. aeruginosa, S. aureus, Candida albi­ The ethanolic extracts of leaf showed significant inhibition and
cans, Aspergillus niger and Aspergillus fumigates (Chaman et al., 2013). indicated excellent anfungal activity against F. moniliforme in extracts.
The studies concluded the ethanolic extracts of C. roseus found to have MIC values of the extract against the tested fungal strains range between
strong properties against C. albicans, P. aeruginosa, and A. niger. Etha­ 25 μg/μl to 50 μg/μl (Kumari and Gupta, 2013). Aqueous ethanolic
nolic extracts of stems and flowers exhibited against S. aureus and extract and its different fractions of leaves of C. roseus have been
A. niger. Chloroform extract of flower has activity against gram-negative examined for their antifungal activity. The highest antifungal activity
bacteria of P. aeruginosa. The aqueous fractions of leaves, stems and was found in the chloroform fraction of C. roseus (Roy and Chatterjee,
flowers were not efficient against bacteria and fungus strains (Chaman 2010). The high antifungal activity has been reported against C. albicans
et al., 2013). The hexane, chloroform, methanol, ethyl-acetate and by methanolic flower extract of C. roseus followed by methanolic leaf
aqueous extracts of root were tested for antibacterial activity against extract and then methanolic stem extract. In vitro extracts showed better
E. coli, Proteus vulgaris, Klebsiella pneumonia, S. aureus, Micrococcus luteus antifungal activity than in vivo extracts due to MZI (30.3 mm ± 0.58a) by
and B. subtilis. The chloroform extracts showed high potential against MIC value is 2.0 mg/mL against B. licheniformis. A similar study of Khalil
almost all tested bacteria species. Gram-negative bacterial strains found (2012) reported the strongest fungal inhibition activity against the
to be less sensitive than Gram-positive bacterial strains (Ethalsha and pathogenic fungi C. albicans (12 mm MZI) using the C. roseus ethanlic
Retna, 2014). The ethyl acetate extracts of different plant parts showed leaf extracts. Ethanolic extract showed high antibacterial while meth­
best among extracts of acetone and methanol of flowers, leaves, stems, anolic extracts reported for high antifungal activity.
and roots for antibiogram followed by acetone and methanol extracts. Bacterial intercellular communication, or quorum sensing (QS),
The extracts of all plant parts were observed the largest antibiogram controls the pathogenesis of many medically important organisms. Anti-
against the bacterial strain B. subtilis followed by K. pneumonia species, QS compounds are known to exist in marine algae and have the ability to
whereas the smallest antibiogram was found against the species of attenuate bacterial pathogenicity. Adonizio et al. (2006) have been
Streptococcus strains. The S. aureus strain was found to be responsive in proposed that the terrestrial plants traditionally used as medicines may
different plant extracts of C. roseus. The ethyl-acetate extract contributed also produce anti-QS compounds. 50 medicinal plants from southern
as the best antibiogram to high solubility which was due to the presence Florida were screened for anti-QS activity using two biomonitor strains,
of bioactives of C. roseus than other solvents used for the study (Sathiya Chromobacterium violaceum and Agrobacterium tumefaciens. Among them
et al., 2008). A study carried out by Kumar et al. (1997) have been re­ only six showed QS inhibition: Conocarpus erectus L. (Combretaceae),
ported screening the seeds of 40 plant species including C. roseus for Chamaecyce hypericifolia (L.) Millsp. (Euphorbiaceae), Callistemon vim­
their antibacterial properties using the strains of B. subtilis, E. coli, inalis (Sol. ex Gaertn.) G. Don (Myrtaceae), Bucida burceras L. (Com­
Pseudomonas cichorii and Salmonella typhimurium, and found the seeds of bretaceae), Tetrazygia bicolor (Mill.) Cogn. (Melastomataceae), and
C. roseus as the most active plants for antibacterial activity. P. cichorii Quercus virginiana Mill. (Fagaceae). This study introduces not only a new
recorded mortlity of 97%, followed by B. subtilis (83%), S. typhimurium mode of action and possible validation for traditional plant use, but also
(69%) and E. coli (55%) bacterial strains. C. roseus extracts antimicrobial a potentially new therapeutic direction for the treatment of bacterial
activity have been carried out and tested using pathogenic bacterial infections.
strains (Bacillus licheniformis, B. subtilis, Azotobacter chroococcum) using
the agar well diffusion method. In vivo and in vitro studies of leaf and in 6.5. Antioxidant activity
vitro study of callus of leaves, fruits and nodal explants in methanol has
been exhibited good antibacterial activity (Naz et al., 2015). The ethanol Antioxidants are protect human body from cancer, inflammatory
extract of leaf found to have showed very strong antibacterial activity disorders and Alzheimer’s diseases (Ethalsha and Retna, 2014). The
against a few human pathogenic microorganisms (S. aureus and E.coli) as different concentrations (100–1000 μg/mL) of chloroform root extracts
well as pathogenic fungi (C. albicans) by the disc diffusion method. The of C. roseus reported to have remarkable antioxidant activity using 2,
100 mg/mL leaf extract found to have the strongest inhibition activity 2-DiPhenyl-1-PicrylHydrazyl radical (DPPH) assay with the IC50
against S. aureus (15 mm MZI) followed by E.coli (11 mm MZI) (Balaa­ (inhibitory concentration). Chloroform extract has higher antioxidant
birami & Patharajan, 2012). potential than standard concentration. The IC50 of C. roseus of extract
In another study, the antimicrobial activity have been reported from has been found at 94.178 μg/mL. Shoots extracts of C. roseus and its
the C. roseus leaf extract by Balaabirami and Patharajan (2012) using fractions (n-hexane, ethyl acetate, methanol, chloroform) exhibited ac­
microorganisms such as E. coli, Klebsiella oxytoca, K. pneumoniae, Proteus tivities related to free radical and percentage inhibition with the linoleic
mirabilis, P. aeruginosa, S. typhimurium, Salmonella paratyphi, Shigella acid for DPPH. The extracts and their fractions have been determined
boydii, S. aureus by disc diffusion method. The result shows that leaf inhibition of peroxidation in linoleic acid (38.4–75.1%) along with good
extracts have the good potential to inhibit the active growth of bacterial DPPH radical IC50, 28.2–119μg/mL The unconventional antioxidant
strains. C. roseus extracts did not exhibit antibacterial activity against activity have been reported in chloroform extracts, it may be presence of
S. aureus, whereas, it was also ineffective against P. aeruginosa. On the volatile compounds in shoot (Widowati et al., 2010). The generally
other hand, C. roseus stem crude extract was inactive against Shigella aqueous and alcoholic extracts are rich source of flavonoids and
boydii. The C. roseus root extract was the most effective against S. typhi phenolic acids therefore, the shoot extracts of C. roseus and fractions in
and S. boydii with the MZI determined 24 mm and 22 mm, respectively. 100% methanol have been found to exhibt strong antioxidant activity,
Raza et al. (2009) have been tested flower extract against Corynebacte­ and the finding showed significant (p < 0.05) variations (Rasool et al.,
rium diphtheriae and found to possess a significant antibacterial activity. 2011). Other reports showed the extracts of C. roseus in methanol have
A study by Shanmugaraju and Bhakyaraj (2016) found that ethanolic the highest DPPH scavenging activity than aqueous extracts (Aadil et al.,
extract exhibited the maximum antibacterial activity in comparison to 2012). Organic acid mixture has been reported from the root extract of
the C. roseus leaf extract prepared in acetone and chloroform. The tested C. roseus and at different concentrations was studied for antioxidant
bacteria S. aureus were susceptible to leaf extracts than tested Escherichia potential by superoxide, DPPH radicals, and nitric oxide. The effect has
coli. The antimicrobial activities of ethanol, acetone and methanol been observed at concentration-dependent up to 19 l g/mL i.e. when

35
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

activity reached its peak. The result indicated that major phytochemical showed zero mortality in comparison to control. The larvicidal activity
constituents of plants are not always responsible for the activity, and against the malarial vector A. stephensi was assayed at various concen­
organic acids mixture with plant extract also have some protective effect trations of leaf extracts and found to be effective for controlling malaria
(Pereira et al., 2010). vectors (Subarani et al., 2013). In another study, the highest larval
A study reported the lyophilised aqueous extract of C. roseus petals mortality was determined in the petroleum ether extract of C. roseus
showed the strongest concentration-dependent antioxidant capacity ef­ against B. thuringiensis due to may be presence of volatile compounds
fects (IC50 at 197 μg-mL), followed by seeds (IC50 at 265 μg-mL) and (Panneerselvam et al., 2013). The larvicidal activity has been evaluated
leaves (IC50 at 447 μg-mL). The extract prepared from flower petals of against larvae with a concentration of 1000 ppm of aqueous extracts.
C. roseus has been found to possess concentration-dependent protective Finding suggested that the larvicidal activity against gram pod borer
effect (Ferreres and Pereira, 2008). The antioxidant property has been Helicoverpa armigera (Lepidoptera: Noctuidae) increased with larval
studied using DPPH and inhibition of malondialdehyde production. The development. The moderating larvicidal effects have been observed in
lowest IC50 for DPPH inhibition found in extract prepared from root all extracts (2.60, 3.94, 7.75, 30.83, 38.53 and 63.34) to 1st, 2nd, 3rd,
hairs (p ≤ 0.01) than leaf callus and root, leaf, of C. roseus (Mardani-­ 4th, 5th and 6th instar of H. armigera larvae after 24 h of exposure to the
Nejad et al., 2016). The water leaf extract had the strongest scavenging extracts. The aqueous leaf extract has been found ED50 values for their
properties of DPPH (22.78 mg/− g), ABTS (35.33 mg/g) and FRAP effect on the larvae of H. armigera and LD50 values were significant at P
(24.18 mg/g), two-fold more of methanol (Pham et al., 2017). The < 0.05. Therefore, the aqueous extract of leaves has potential to be use
ethanol and ethyacetate extracts showed lower antioxidant capacity like biopesticide for H. armigera management (Alaguchamy and Jaya­
than H2O over sixfold and ninefold lower, respectively. The drying kumararaj, 2015). The pupicidal activity of leaf extract was reported
procedure has been reported significantly affected the antioxidant 73.33, 49.33, 33.33, 28.00 and 17.33 percent mortality was observed
properties of C. roseus. The vacuum drying (VD50) at 50 ◦ C was suitable when pre-pupae were treated with 2.0, 1.5, 1.0, 0.5 and 0.1%, respec­
for the leaf whereas and the infrared drying (IRD35) at 35 ◦ C was sug­ tively in the pre-pupal stage of Spodoptera litura using topical applica­
gested for stem, root, and flower due to phenolic contents (Pham et al. tion method. The leaf alcoholic extracts are the good results for
(2018). The antioxidant properties were studied using the ethanol leaf treatment against larvae and pupae (Sandey and Summarwar, 2016).
extracts of Piper betle, Datura petandra, Curcuma mangga and C. roseus
following DPPH assays. The IC50 values of D. petandra (4.74) and P. betle 6.8. Other activities
(5.49) found to be lowerest than C. roseus (102.96), C. mangga (277.79)
extracts. These findings indicated P. betle and D. petandra extracts are Compound 52 has been used for Alzheimer’s disease (AD) and de­
better antioxidants in comparison to C. roseus and C. manga plant species mentia populations. This active compound has been reported to be
(Widowati et al., 2011; Verma et al., 2013). The significant effect was potent at the doses up to 60 mg/d in clinical trials of dementia and stroke
also reported in C. roseus with the highest ORAC values (2.85 mg of (Lamar et al., 1988; Krieglstein and Rischke, 1991; Lakics et al., 1995;
GAE/g of fresh weight) (Zhao et al., 2001). Orviský et al., 1997; Gabryel et al., 2002; Manda et al., 2015). The
ethanol extract of the flower of C. roseus has been more strongly stim­
6.6. Hepatoprotective activity ulating accelerated wound healing activity compared to controls (Nayak
et al., 2007). Tensile strength has been enhanced by hydroxyproline
Associated liver diseases are considered as the most serious health content and the hypolipidemic active principles due to the influence of
problems, as this organ is responsible for metabolism and excretion, and ethanol extract (Senbagalakshmi et al., 2017). The hydroalcoholic or
frequently exposed to various diverse xenobiotics and therapeutic dichloromethane-methanol extracts of leaves and stem and roots have
agents. Ahmed and Rao (2013) have been reported comparative hep­ shown significant effects in hypotensive (Ara et al., 2009). Hoskeri et al.
atoprotective activities of C. rosea, Brassica oleracea, and Melia azedarach (2011) studied in-vitro anthelminthic activity using whole plant parts of
leaves ethanolic extracts with simvastatin induced hepatotoxicity in C. roseus in ethanol and water extracts. Different concentrations of
rats. Hepatotoxicity induced by simvastatin at a dose of 20 mg/kg for ethanol extract was used for study against P. posthuma. The efficient
one month in rats, whereas protected by M. azedarach, C. rosea, and paralysis effect (6.67mins) was observed in the ethanol extract at 200
B. oleracea (300 mg/kg and 500 mg/kg). Hepatotoxicity-induced rats mg/mL than other extracts. Similarly, ethanol extract with dose at 250
found to have significant changes in biochemical parameters such as mg/mL showed significant anthelmintic activity with a lead time of
enhanced serum glutamic pyruvic transaminase (SGPT), serum 46.33 mins (Hoskeri et al., 2011). Sadique and Hashim (2019) investi­
glutamic-oxaloacetic transaminase (SGOT), alkaline phosphatase (ALP), gated the antiangiogenesis activity of C. roseus with ethyl acetate, which
serum bilirubin, and decrease in proteins content. These biochemical exhibited highly potent antiangiogenesis activity. The efficacy of this
parameters were restored in rats treated with extracts. Finding showed activity may be due to the presence of flavonoids because the ethyl ac­
ethanolic extracts of M. azedarach at a dose of 300 mg/kg and B. oleracea etate fraction was flavonoid-rich. The antimutagenic activity was
at a dose of 500 mg/kg found to have potent hepatoprotective properties commonly reported in anticancer drugs from medicinal plants and they
in comparison to C. rosea extract. Histopathological studies of liver were contain direct or indirect antimutagens (Banerjee and Mumtaz, 2020).
also reported which indicates further confirmation of the hep­ The antimutagenic actions of triterpenes, hydrocarbons and an alkaloid
atoprotective effect of the extracts. isolated from C. roseus has been reported (Inte et al., 1998; Mishra and
Verma, 2017).
6.7. Larvicidal and pupicidal property The antidiarrheal effect of C. roseus was studied with extracts at 200
and 500 mg/kg doses in castor oil pretreated diarrheal rats. Standard
C. roseus plants larvicidal as well as pupicidal properties have been drug loperamide and atropine sulfate ware used during two experi­
explored by many researchers (Kuppusamy et al., 2009; Sandey and ments. The ethanolic extract has been reported as a dose-dependent
Summarwar, 2016: Pavunraj et al., 2017). The methanol, aqueous and inhibition of castor oil-induced diarrhoea. The most significant differ­
ethyl-acetate of leaf extract have been tested against malaria and fila­ ence was observed at 200 mg/kg (P < 0.05) than at 500 mg/kg (P <
riasis vectors such as 4th instar larvae/pupae of Anopheles stephensi and 0.001) in the extract with the negative control, which suggested a sig­
Culex quinquefasciatus. Results showed that the lethal doses of LC50 and nificant antidiarrheal effect (Hassan et al., 2011).
LC90 at 50% and 90% of the used samples of A. stephensi, C. quinque­ The ethanol extract of leaf has been reported for antiplasmodial ac­
fasciatus which were killed after post treatment of 24 and 48 h. In tivity against both chloroquine-sensitive Pf3D7 and chloroquine resis­
aqueous extract, the mortality of larval/pupal was observed after 24 and tant PfINDO strains of Plasmodium falciparum grown in human red blood
48 h, whereas samples treated with ethyl-acetate and methanol extracts cell cultures with IC50 11–20 μg/mL (Singh et al., 2015). Leaves extract

36
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

can be a good source for synthesis of nanoparticles which shows anti­ was observed in the concentration of 5% as 76.67 ± 21.34% and in the
plasmodial activity against P. falciparum (Ponarulselvam et al., 2012). control there was no mortality (Kumar et al., 2011).
Bisindole alkaloids have been used in clinic applications due to their
7. Clinical applications antitumor effects. Toxicities of bisindole alkaloids are significantly
different despite similar structures. However, the structure-toxicity re­
Structures of active leads and drugs that have been identified from lationships of bisindole alkaloids remain unclear. Wang et al. (2017)
plant parts of C. roseus and currently used for the treatment of various have evaluated the toxic effects of bisindole alkaloids using Tetrahymena
cancers are presented in 3A, 3B, 4A, 4B. Compound vincaleukoblastine, thermophile BF5 (T.t. BF5) which could dynamically monitor the growth
VelbeR has been isolated in 1960, used in the treatment of Hodgkin’s of T.t. BF5. The IC10 results showed the relative toxicity of bisindole
disease, non-Hodgkin lymphomas, testiscarcinomas and sometimes alkaloids was compound 112 (105.0 ± 6.3 μg/mL) > compound 121
against breast cancer and chorio-carcinomas (Kris et al., 1985; Levêque (135.1 ± 5.1 μg/mL) > compound 111 (215.5 ± 4.7 μg/mL) > com­
and Jehl, 2007). Compound 112 (leurocristine, OncovinR) is oxidized pound 143 (314.2 ± 5.9 μg/mL). Structure-toxicity relationships anal­
form of compound 111 and marketed and commercialized in 1963, and ysis of these alkaloids indicated that the acylamide at C-3 position and
used against acute leukemia, Hodgkin’s disease, non-Hodgkin lym­ hydroxyl at C-4 position might contribute to more toxicity. A long
phomas, Wilm’s tumors in children, rhabdomyosarcomas and breast conjugation effect appearing in bisindole alkaloids could increase the
cancer (Kaplan et al., 1986; Parthasarathy et al., 2020). Anhydrovin­ toxicity when the N1 position was substituted with the formyl group.
blastine was recommended for use as an antineoplastic agent in the In humans, the dose-limiting toxicity of compound 112 is a periph­
treatment of cervical and lung cancer (Quan et al., 2019; Wiernikowski eral neuropathy. The effects are believed to be limited to peripheral
and Bernhardt, 2020). Some C. roseus alkaloids marketed and nerves because compound112 does not penetrate the blood–brain bar­
commercialized in 1957 for the treatment of hypertension as pharma­ rier. Compound 112 was interrupts microtubular function, resulting in
ceuticals include compound 80 (HydroserpanR, LamuranR) (Senbaga­ an inability to maintain axonal membrane integrity (Grush and Morgan,
lakshmi et al., 2017). This is often considered as the major drug of 1979; Bradfield et al., 2015; Li et al., 2020). Compound 111 is typically
R. serpentina (Kumar et al., 2016a, 2016b). The most active bisindole well tolerated, with rare GI or myelosuppressive side effects. Extrava­
alkaloids consist of a plumeran (compound 27) and an ibogan (com­ sation of compound 111 may cause significant tissue irritation and
pound 55) moiety (Jacobs et al., 2004; Das and Sharangi, 2017). Besides cellulitis; therefore the drug must be given through a clean IV puncture
alkaloids, other secondary metabolites have been isolated from C. roseus, (King and Boder, 1979).
including monoterpenoid glucosides (loganin, secologanin, sweroside, The ethnobotanical surveys did not mention any toxicity associated
deoxyloganin, dehydrologanin), steroids (catasteron, brassinolides), with the use of this plant. Still, there may be inadequate evidence to
phenolics, flavonoids, and anthocyanins showed considerable potential overrule the toxicity and safety of this plant. Hence, further in-depth in
can be used for further study (Federico et al., 2009; Kabesh et al., 2015; vitro and in vivo studies for toxicity must be considered.
Zhang et al., 2018).
9. Conclusion
8. Toxicity
This communication provides an updated and critically analyzed
The toxicity of ethanolic, methanolic and aqueous extracts from summary concerning the botany, traditional uses, phytochemistry,
plant parts of C. roseus have been inadequately studied (James et al., separation, analysis techniques and pharmacology and toxicity aspects
2007). Some studies had toxicity for extracts and isolated compounds at of parts of C. roseus. Traditional uses of C. roseus are confirmed by
above the optimized doses. Methanolic extract of leaf studied for liver different studies pertaining to pharmacological functions such as anti­
and kidney functions in Dawely rats using 0.1, 0.5 and 1.0 g/kg. Dose at microbial, antifertility effect, antioxidant, antidiabetic, hep­
1.0 g/kg causes side effects such as mortality and diarrhoea but 0.1 g/kg atoprotective, larvicidal/pupicidal effect in different extracts especially
was safe without causing effects on liver and kidney functions (Kevin alcoholic extracts of leaves and its fractions. The phytochemical in­
et al., 2012). Aqueous extract studied for acute toxicity in male Wistar vestigations revealed that bisindole and MIAs were the two major classes
rats using at 1000 and 5000 mg/kg body weight. The ethanolic extract of of phytoconstituents that contribute to the various biological effects and
leaf causes toxicity doses at higher than 300 mg/kg BW can produce localized in leaves. The phenolic acids and flavonoids are responsible for
signs of biochemical and histopathological toxicity in liver, kidney and antioxidant properties whereas antimicrobial activity could be due to
heart in wistar albino rats. The dose of 2000 mg did not cause mortality. presence of terpenes, and volatile compounds which are reported as a
SGOT, SGPT, creatinine phosphokinase, LDH, urea and creatinine were minor class of compounds. The recent clinical studies of the most active
increased in 300 mg/kg BW and 2000 mg/kg BW doses (Vutukuri et al., phytochemicals, compounds 111, 112 and 121 are employed for the
2017). Therefore, ethanolic methanolic and aqueous aqueous extracts of treatment of various types of chemotherapy due to their cell toxicity.
leaves found to be safety in dose dependent manner. The toxicological researches of C. roseus are expected to provide the
The study recommends the use of lower concentrations than 1000 important data which may contribute to drug discovery having
mg/kg in order to increase the safety margin. Safety measures that improved and intriguing pharmacology aspects. We hope these sum­
include monitoring of the vital serum enzyme and hematological pa­ marized data will be a prevalent aspect of future drug discovery
rameters are recommended when this extract is being administered. research.
Comprehensive screening for possible toxicity on sub-acute, sub-chronic
and chronic levels was also recommended (Kabubii et al., 2015). A study CRediT authorship contribution statement
has been reported the poisoning effects of C. roseus in sheep during long
time grazing of this plant. This study demonstrates that C. roseus can Sunil Kumar: conceived the idea, designed, collected, compiled, and
cause toxicosis in sheep resulting in mortality probably due to dissem­ prepared the manuscript. and, wrote the article, prepared tables., drew
inated intravascular coagulation and circulatory disturbances related to the chemical structures, designed the graphical abstract, using Chem­
diarrhoea and dehydration. In conclusion, results of this study suggested Draw pro software, and cited the references as per JEP guidelines, hel­
that the diagnosis of this poisoning depends upon a combination of ped in editing the manuscript. Bikarma Singh: conceived the idea,
anamnesis, clinical signs, laboratory parameters and pathological ex­ designed, collected, compiled, and prepared the manuscript, helped in
amination (Aydogan et al., 2015). The acetone extract of root has been editing the manuscript. Ramesh Singh: helped in editing the
affected on the larval mortality, fecundity and fertility of Spodoptera manuscript.
litura (Fabricius) as concentration increases. The maximum mortality

37
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Declaration of competing interest Hybrid Triple Quadrupole-Linear Ion Trap Mass Spectrometry
UPLC Ultra-Performance Liquid Chromatography
The authors declare no conflict of interest. UV–vis UltraViolate Visible
VLDL-c Very Low Density Lipoprotein
Acknowledgments XRD X-Ray Diffraction Patterns

Authors are thankful to Principal, Ma. Kanshiram Government De­ References


gree College, Ninowa, Farrukhabad, and Director CSIR NBRI Lucknow
for their kind supports and encouragement. Aadil, K.R., Barapatre, A., Rathore, N., Pottam, S., Jha, H., 2012. Comparative study of
in-vitro antioxidant and antidiabetic activity of plant extracts of Acacia arabica,
Murraya koeingii, Catharanthus roseus and Rouwolfia serpentina. Int. J. Phytomed. 4,
Abbreviations 543–551.
Adekomi, A.D., 2020. Madagascar periwinkle (Catharanthus roseus) enhances kidney and
liver functions in Wistar rats. Eur. J. Anat. 14, 111–119.
AI Ambient Ionization
Adonizio, A.L., Downum, K., Bennett, B.C., Mathee, K., 2006. Anti-quorum sensing
ALP Alanine Phosphatase activity of medicinal plants in southern Florida. J. Ethnopharmacol. 105, 427–435.
ESI Electrospray Ionization Aerts, R.J., Gisi, D., De Carolis, E., De Luca, V., Baumann, T.W., 1994. Methyl jasmonate
HDL-c High-Density Lipoprotein Cholesterol vapor increases the developmentally controlled synthesis of alkaloids in
Catharanthus and Cinchona seedlings. Plant J. 5, 635–643.
HPLC-ESI-MS/MS High-Performance Liquid Chromatography- Ahmad, N.H., Rahim, R.A., Mat, I., 2010. Catharanthus roseus aqueous extract is cytotoxic
Electrospray Ionization Tandem Mass Spectrometry to Jurkat leukaemic T-cells but induces the proliferation of normal peripheral blood
LDL-c Low-Density Lipoprotein Cholesterol mononuclear cells. Trop. Life Sci. Res. 21, 101–113.
Ahmed, M.F., Kazim, S.M., Ghori, S.S., Mehjabeen, S.S., Ahmed, S.R., Ali, S.M.,
SEM Scanning Electron Microscopy Ibrahim, M., 2010. Antidiabetic activity of Vinca rosea extracts in alloxan-induced
SGOT Serum Glutamate Oxaloacetate Transaminase diabetic rats. Internet J. Endocrinol. 2010, 1–6.
AIHR Adrenaline-Induced Hypertensive Rats Ahmed, M.F., Rao, A.S., 2013. Comparative hepatoprotective activities of selected Indian
medicinal plants. Global J. Med. Res. 12, 15–22.
APCI Atmospheric Pressure Chemical Ionization Akhtar, M.A., Rashid, M., Wahed, M.I.I., Islam, M.R., Shaheen, S.M., Islam, M.A.,
APP Atmospheric Pressure Photoionization Amran, M.S., Ahmed, M., 2007. Comparison of long-term antihyperglycemic and
BHA Butylated HydroxyAnisol hypolipidemic effects between Coccinia cordifolia Linn. and Catharanthus roseus Linn.
in alloxan-induced diabetic rats. RJMMS 2, 29–34.
BW Body weight
Alaguchamy, N., Jayakumararaj, R., 2015. Larvicidal effect of Catharanthus roseus L (G)
CE-MS Capillary Electrophoresis–Mass Spectrometry Don. aqueous leaf extracts on the larvae of Helicoverpa armigera (Hübner). Int. J.
CNS Central Nervous System Zool. Appl. Biosci. 3, 10–14.
Alam, M.M., Naeem, M., Khan, M.M.A., Uddin, M., 2017. Vincristine and Vinblastine
CVD CardioVascular Diseases
Anticancer Catharanthus Alkaloids: Pharmacological Applications and Strategies for
DPPH 2, 2-DiPhenyl-1-PicrylHydrazyl radical Yield Improvement. In: Catharanthus Roseus, pp. 277–307. Springer, Cham. New
EDX Energy Dispersive X-Ray York City.
FBG Fasting Blood Glucose Alam, P., Sharaf-Eldin, M.A., 2016. Limited production of plant derived anticancer drugs
vinblastine and vincristine. Planta Med. 82 (4), 10.1055/s-0036-1578706.
FRAP Ferric Reducing Antioxidant Power Alcover, C.F., Bernadat, G., Kabran, F.A., Le Pogam, P., Leblanc, K., Fox Ramos, A.E.,
GC-MS Gas Chromatography Mass Spectrometry Gallard, J.F., Mouray, E., Grellier, P., Poupon, E., Beniddir, M.A., 2020. Molecular
HPLC High-Performance Liquid Chromatography networking reveals serpentinine-related bisindole alkaloids from Picralima nitida, a
previously well-investigated species. J. Nat. Prod. 83, 1207–1216.
HPTLC High-Performance Thin Layer Chromatography Allorge, L., Phillipson, P.B., Razakamalala, R., 2015. Catharanthus makayensis L.
IC50 50% Maximal Inhibitory Concentration Allorge, phillipson & Razakamal. (Apocynaceae), a new species from Madagascar.
ITAS and Indian traditional Ayurvedic system Candollea 70, 61–67.
Al-Quteimat, O.M., 2020. Vincristine shortage: what other options we have? Am. J. Clin.
LC-MS Liquid Chromatography Mass Spectrometry Oncol. 43, 146–147.
MALDI-TOF Matrix Assisted Laser Desorption with Time-of-Flight Al-Shaqha, W.M., Khan, M., Salam, N., Azzi, A., Chaudhary, A.A., 2015. Anti-diabetic
MIAs Monoterpene Indole Alkaloids potential of Catharanthus roseus Linn. and its effect on the glucose transport gene
(GLUT-2 and GLUT-4) in streptozotocin induced diabetic wistar rats. BMC
MIC Minimum Inhibitory Concentration Complement. Altern. Med. Times 15, 379–387.
MZI Minimum Zone Inhibition Andrade, S.A.L., Malik, S., Sawaya, A.C.H.F., Bottcher, A., Mazzafera, P., 2013.
MRM Multiple Reaction Monitoring Association with arbuscular mycorrhizal fungi influences alkaloid synthesis and
accumulation in Catharanthus roseus and Nicotiana tabacum plants. Acta Physiol.
mRNA Messenger RiboNucleic Acid
Plant 35, 867–880.
MS Mass Spectrometry Ara, N., Rashid, M., Amran, S., 2009. Comparison of hypotensive and hypolipidemic
MZI Maximum Zone of Inhibition effects of Catharanthus roseus leaves extract with atenolol on adrenaline induced
ORAC Oxygen Radical Absorbance Capacity hypertensive rats. Pak. J. Pharm. Sci. 22, 267–271.
Arora, R., Malhotra, P., Mathur, A.K., Mathur, A., Govil, C.M., Ahuja, P.S., 2010.
PCR Polymerase Chain Reaction Anticancer alkaloids of Catharanthus roseus: transition from traditional to modern
PCV Packed Cell Volume medicine. Herbal Medicine: A Cancer Chemopreventive and Therapeutic
PDA Photo Diode Array Perspective. Jaypee Brothers Medical Publishers Pvt. Ltd, New Delhi, pp. 292–310.
New Delhi, India.
RAPD Random Amplified Polymorphic Aruljothi, B., Samipillai, S.S., 2016. Antidiabetic activity of Catharanthus roseus in
DNA, rDNA Ribosomal DeoxyriboNucleic Acid alloxan induced diabetic rats. Int. J. Modn. Res. Revs. 4, 1121–1124.
SGPT Serum Glutamate Pyruvate Transaminase Aruna, M.S., Prabha, M.S., Priya, N.S., Nadendla, R., 2015. Catharanthus roseus:
ornamental plant is now medicinal boutique. J. Drug Deliv. Ther 5, 1–4.
SIM Single Ion Monitoring Aslam, J., Khan, S.H., Siddiqui, Z.H., Fatima, Z., Maqsood, M., Bhat, M.A., Nasim, S.A.,
STZ StrepToZotocin Ilah, A., Ahmad, I.Z., Khan, S.A., Mujib, A., 2010. Catharanthus roseus (L.) G. Don. An
TC Total Cholesterol important drug: it’s applications and production. Pharm. Glob 4, 1–16.
Aslam, J., Mujib, A., Nasim, S.A., Sharma, M.P., 2009. Screening of vincristine yield in ex
TCM traditional Chinese medicine, vitro and in vitro somatic embryos derived plantlets of Catharanthus roseus L. (G)
TEAC Trolox Equivalent Antioxidant Capacity Don. Sci. Hortic 119, 325–329.
TG TriGlyceride, Aslam, J., Mujib, A., Sharma, M.P., 2011. Influence of freezing and non-freezing
temperature on somatic embryogenesis and vinblastine production in Catharanthus
TGA Cholesterol total, triglyceride,
roseus (L.) G. Don. Acta Physiol. Plant 33, 473–480.
TOSC Total Oxyradical Scavenging Capacity Ataei-Azimi, A., Hashemloian, B.D., Ebrahimzadeh, H., Majd, A., 2008. High in vitro
TSI Thermospray Ionization production of ant-canceric indole alkaloids from periwinkle (Catharanthus roseus)
TSP-MS Thermospray- Mass Spectrometry tissue culture. Afr. J. Biotechnol. 7, 2834–2839.
Auriola, S., Ranta, V.P., Naaranlahti, T., Lapinjoki, S.P., 1989. Thermospray liquid
UHPLC-ESI-QqQLIT- MS/MS Ultra-High-Performance Liquid chromatography-mass spectrometric analysis of Catharanthus alkaloids.
Chromatography Coupled With Electrospray Ionization J. Chromatogr. Abstr. 474, 181–185.

38
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Aydogan, A., Sezer, K., Ozmen, O., Haligur, M., Albay, M.K., 2015. Clinical and Das, S., Sharangi, A.B., 2017. Madagascar periwinkle (Catharanthus roseus L.): diverse
pathological investigations of accidental Catharanthus roseus toxicity in sheep. Israel medicinal and therapeutic benefits to humankind. J. Pharmacogn. Phytochem. 6,
J. Vet. Med. 70, 51–56. 1695–1701.
Ayob, F.W., Simarani, K., Zainal Abidin, N., Mohamad, J., 2017. First report on a novel N De Pinho, P.G., Gonçalves, R.F., Valentão, P., Pereira, D.M., Seabra, R.M., Andrade, P.B.,
igrospora sphaerica isolated from Catharanthus roseus plant with anticarcinogenic Sottomayor, M., 2009. Volatile composition of Catharanthus roseus (L.) G. Don using
properties. Microb. Biotechnol 10, 926–932. solid-phase microextraction and gas chromatography/mass spectrometry.
Baig, M.M.V., Ingle, S.S., 2021. Induction of callus in Catharanthus roseus (L.) G. Don and J. Pharmaceut. Biomed. Anal. 49, 674–685.
screening for secondary metabolites in callus cells. Eur. J. Mol. Clin. Med. Times 7, Digvijay, S., Shashi, P.R., Suchi, S., Kumar, R.S., Raghavendra, M., Sushil, K., 2004.
2911–2914. Simultaneous quantification of some pharmaceutical Catharanthus roseus leaf and
Balaabirami, S., Patharajan, S., 2012. In vitro antimicrobial and antifungal activity of root terpenoid indole alkaloids and their precursors in single runs by reversed-phase
Catharanthus roseus leaves extract against important pathogenic organisms. Int. J. liquid chromatography. J. AOAC Int. 87, 1287–1296.
Pharm. Pharmaceut. Sci. 4, 487–490. Don, G., 1999. Catharanthus roseus. In: Ross, I.A. (Ed.), Medicinal Plants of the World.
Banerjee, A., Mumtaz, S.M.F., 2020. In vitro antimutagenic activity of Ipomoea Springer Science+Business Media New York, Humana Press, pp. 109–118.
staphylina. Bangladesh J. Pharmacol 15, 41–43. Doshi, G.M., Matthews, B.D., Chaskar, P.K., 2018. Gas chromatography-mass
Barkat, M.A., Abul, H., Rahman, M.A., 2017. Agricultural, pharmaceutical, and spectroscopy studies on ethanolic extract of dried leaves of Catharanthus roseus.
therapeutic interior of Catharanthus roseus (L.) G. Don. In: Catharanthus Roseus. Asian J. Pharm. Clin. Res. 11, 336–340.
Springer, Cham, pp. 71–100. Dutta, A., Batra, J., Pandey-Rai, S., Singh, D., Kumar, S., Sen, J., 2005. Expression of
Barrales-Cureño, H.J., Reyes, C.R., García, I.V., Valdez, L.G.L., De Jesús, A.G., Ruíz, J.A. terpenoid indole alkaloid biosynthetic pathway genes corresponds to accumulation
C., Herrera, L.M.S., Caballero, M.C.C., Magallón, J.A.S., Perez, J.E., Montoya, J.M., of related alkaloids in Catharanthus roseus (L.) G. Don. Planta 220, 376–383.
2019. Alkaloids of pharmacological importance in catharanthus roseus. In alkaloids- El-Domyati, F.M., Abdel Azeiz, A.Z., Atef, A., Elhalwagi, A., Sabir, J.S.M., Ramadan, A.
their importance in nature and human life. IntechOpen. https://doi.org/10.5772/ M., Gadalla, N.O., Edris, S., Baeshen, M.N., Hajrah, N.H., Al-Kordy, M.A., 2014.
intechopen.82006. Functional genomic analysis for overproduction of key terpenoid indole alkaloids in
Barthe, L., Ribet, J.P., Pélissou, M., Degude, M.J., Fahy, J., Duflos, A., 2002. mature leaf of Catharanthus roseus. Life Sci. 11, 1313–1320.
Optimization of the separation of Vinca alkaloids by nonaqueous capillary El-Ghazaly, G., 1990. Development of pollen grains of Catharanthus roseus
electrophoresis. J. Chromatogr. A 968, 241–250. (Apocynaceae). Rev. Palaeobot. Palynol. 64, 165–174.
Batra, J., Dutta, A., Singh, D., Kumar, S., Sen, J., 2004. Growth and terpenoid indole El-Sayed, M., Choi, Y.H., Frederich, M., Roytrakul, S., Verpoorte, R., 2004. Alkaloid
alkaloid production in Catharanthus roseus hairy root clones in relation to left-and accumulation in Catharanthus roseus cell suspension cultures fed with stemmadenine.
right-termini-linked Ri T-DNA gene integration. Plant Cell Rep. 23, 148–154. Biotechnol. Lett. 26, 793–798.
Benyammi, R., Paris, C., Khelifi-Slaoui, M., Zaoui, D., Belabbassi, O., Bakiri, N., Meriem Ethalsha, P., Retna, A.M., 2014. Evaluation of antioxidant potential and antibacterial
Aci, M., Harfi, B., Malik, S., Makhzoum, A., Desobry, S., 2016. Screening and kinetic activity of crude extracts Catharanthus roseus. Int. J. Pharma Sci. Res. 5, 3490–3495.
studies of catharanthine and ajmalicine accumulation and their correlation with Falcão, M.A., Scopel, R., Almeida, R.N., do Espirito Santo, A.T., Franceschini, G.,
growth biomass in Catharanthus roseus hairy roots. Pharm. Biol. 54, 2033–2043. Garcez, J.J., Vargas, R.M., Cassel, E., 2017. Supercritical fluid extraction of
Bertaccini, A., 2007. Phytoplasmas: diversity, taxonomy, and epidemiology. Front. vinblastine from Catharanthus roseus. J. Supercrit. Fluids 129, 9–15.
Biosci. 12, 673–689. Favali, M.A., Musetti, R., Benvenuti, S., Bianchi, A., Pressacco, L., 2004. Catharanthus
Bhadra, R., Vani, S., Shanks, J.V., 1993. Production of indole alkaloids by selected hairy roseus L. plants and explants infected with phytoplasmas: alkaloid production and
root lines of Catharanthus roseus. Biotechnol. Bioeng. 41, 581–592. structural observations. Protoplasma 223, 45–51.
Bihani, T., 2021. Plumeria rubra L.– A review on its ethnopharmacological, Favretto, D., Piovan, A., Filippini, R., Caniato, R., 2001. Monitoring the production yields
morphological, phytochemical, pharmacological and toxicological studies. of vincristine and vinblastine in Catharanthus roseus from somatic embryogenesis.
J. Ethnopharmacol. 264 (113291) https://doi.org/10.1016/j.jep.2020.113291. Semiquantitative determination by flow-injection electrospray ionization mass
Bisla, G., Choudhary, S., Chaudhary, V., 2014. Evaluation of the nutritive and spectrometry. Rapid Commun. Mass Spectrom. 15, 364–369.
organoleptic values of food products developed by incorporated Catharanthus roseus Federico, M., Luminari, S., Iannitto, E., Polimeno, G., Marcheselli, L., Montanini, A., La
(Sadabahar) fresh leaves explore their hypoglycemic potential. Sci. World J. 2014, Sala, A., Merli, F., Stelitano, C., Pozzi, S., Scalone, R., 2009. ABVD compared with
1–6. BEACOPP compared with CEC for the initial treatment of patients with advanced
BlaskÓ, G., Cordell, G.A., 1990. Isolation, structure elucidation, and biosynthesis of the Hodgkin’s lymphoma: results from the HD2000 Gruppo Italiano per lo Studio dei
bisindole alkaloids of Catharanthus. Alkaloids Chem. Pharmacol. 37, 1–76 Linfomi Trial. J. Clin. Oncol. 27, 805–811.
(Academic Press). Fernández-Pérez, F., Almagro, L., Pedreño, M.A., Gomez Ros, L.V., 2013. Synergistic and
Bradfield, S.M., Sandler, E., Geller, T., Tamura, R.N., Krischer, J.P., 2015. Glutamic acid cytotoxic action of indole alkaloids produced from elicited cell cultures of
not beneficial for the prevention of vincristine neurotoxicity in children with cancer. Catharanthus roseus. Pharm. Biol. 51, 304–310.
Pediatr. Blood Canc. 62, 1004–1010. Ferreres, F., Figueiredo, R., Bettencourt, S., Carqueijeiro, I., Oliveira, J., Gil-
Brun, G., Bessière, J.M., Dijoux-Franca, M.G., David, B., Mariotte, A.M., 2001. Volatile Izquierdo, A., Pereira, D.M., Valentão, P., Andrade, P.B., Duarte, P., Barceló, A.R.,
components of Catharanthus roseus (L.) G. Don (Apocynaceae). Flavour Fragrance J. 2011. Identification of phenolic compounds in isolated vacuoles of the medicinal
16, 116–119. plant Catharanthus roseus and their interaction with vacuolar class III peroxidase: an
Chaman, S., Sharma, G., Anil, S., Reshi, K., 2013. Study of antimicrobial properties of H2O2 affair? J. Exp. Bot. 62, 2841–2854.
Catharanthus roseus by agar well diffusion method. Int. Res. J. Pharmaceut. Appl. Sci. Ferreres, F., Pereira, D.M., Valentao, P., Andrade, P.B., Seabra, R.M., Sottomayor, M.,
5, 65–68. 2008. New phenolic compounds and antioxidant potential of Catharanthus roseus. J
Chen, Q., Li, N., Zhang, W., Chen, J., Chen, Z., 2011. Simultaneous determination of Agr. Food Chem. 56, 9967–9974.
vinblastine and its monomeric precursors vindoline and catharanthine in Ferreres, F., Pereira, D.M., Valentão, P., Oliveira, J.M., Faria, J., Gaspar, L.,
Catharanthus roseus by capillary electrophoresis–mass spectrometry. J. Separ. Sci. 34, Sottomayor, M., Andrade, P.B., 2010. Simple and reproducible HPLC–DAD–ESI-MS/
2885–2892. MS analysis of alkaloids in Catharanthus roseus roots. J. Pharmaceut. Biomed 51,
Chen, Q., Zhang, W., Zhang, Y., Chen, J., Chen, Z., 2013. Identification and 65–69.
quantification of active alkaloids in Catharanthus roseus by liquid Filippini, R., Caniato, R., Piovan, A., Cappelletti, E.M., 2003. Production of anthocyanins
chromatography–ion trap mass spectrometry. Food Chem. 139, 845–852. by Catharanthus roseus. Fitoterapia 74, 62–67.
Choi, Y.H., Yoo, K.P., Kim, J., 2002. Supercritical fluid extraction and liquid Fitrianto, N., Proklamasiningsih, E., Muljowati, J.S., 2020. Phytochemical diversity and
chromatography-electrospray mass analysis of vinblastine from Catharanthus roseus. antimicrobial properties of methanol extract of several cultivars of Catharanthus
Chem. Pharm. Bull. 50, 1294–1296. roseus using GC-MS. Biodiv. J. Biol. Diver 21, 1332–1344.
Chu, I.H., Bodnar, J.A., Bowman, R.N., White, E.L., 1997. Determination of vincristine Frasci, G., Lorusso, V., Panza, N., Comella, P., Nicolella, G., Bianco, A., De Cataldis, G.,
and vinblastine in Catharanthus roseus plants by high performance liquid Iannelli, A., Bilancia, D., Belli, M., Massidda, B., 2000. Gemcitabine plus vinorelbine
chromatography/electrospray ionization mass spectrometry. J. Liq. Chromatogr. versus vinorelbine alone in elderly patients with advanced non–small-cell lung
Relat. Technol. 20, 1159–1174. cancer. J. Clin. Oncol.18 2529–2536.
Chung, I.M., Ahmad, A., Ali, M., Lee, O.K., Kim, M.Y., Kim, J.H., Yoon, D.Y., Peebles, C. Fukuyama, T., Ohashi-Kaneko, K., Watanabe, H., 2015. Estimation of optimal red light
A., San, K.Y., 2009. Flavonoid glucosides from the hairy roots of Catharanthus roseus. intensity for production of the pharmaceutical drug components, vindoline and
J. Nat. Prod. 72, 613–620. catharanthine, contained in Catharanthus roseus (L.) G. Don. Environ. Control. Biol.
Cui, M., Song, F., Zhou, Y., Liu, Z., Liu, S., 2000. Rapid identification of saponins in plant 53, 217–220.
extracts by electrospray ionization multi-stage tandem mass spectrometry and liquid Gabryel, B., Adamek, M., Pudełko, A., Małecki, A., Trzeciak, H.I., 2002. Piracetam and
chromatography/tandem mass spectrometry. Rapid Commun. Mass Spectrom. 14, vinpocetine exert cytoprotective activity and prevent apoptosis of astrocytes in vitro
1280–1286. in hypoxia and reoxygenation. Neurotoxicology 23, 19–31.
da Silva e Silva, J.V., Cordovil Brigido, H.P., Oliveira de Albuquerque, K.C., Miranda Gajalakshmi, S., Vijayalakshmi, S., Devi, R.V., 2013. Pharmacological activities of
Carvalho, J., Ferreira Reis, J., Vinhal Faria, L., Coelho-Ferreira, M., Silveira, F.T., da Catharanthus roseus: a perspective review. Int. J. Pharma. Bio. Sci.4 431–439.
Silva Carneiro, A., Percário, S., do Rosário Marinho, A.M., 2019. Flavopereirine—an Gomber, S., Dewan, P., Chhonker, D., 2010. Vincristine induced neurotoxicity in cancer
alkaloid derived from geissospermum vellosii-presents leishmanicidal activity in patients. Indian J. An. Pediatr. 77, 97–100.
vitro. Molecules 24, 785. Govindasamy, C., Srinivasan, R., 2012. In vitro antibacterial activity and phytochemical
Dai, Y., Rozema, E., Verpoorte, R., Choi, Y.H., 2016. Application of natural deep eutectic analysis of Catharanthus roseus (Linn.) G. Don. Asian Pac. J. Trop. Biomed 2,
solvents to the extraction of anthocyanins from Catharanthus roseus with high 155–158.
extractability and stability replacing conventional organic solvents. J. Chromatogr. A Grush, O.C., Morgan, S.K., 1979. Folinic acid rescue for vincristine toxicity. Clin. Toxicol.
1434, 50–56. 14, 71–78.

39
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Gumbi, B.P., Moodley, B., Birungi, G., Ndungu, P.G., 2019. Target, suspect and non- Kayesth, S., Shazad, M., Kumar, S., Gupta, K.K., 2017. Effect of ethanol extract of
target screening of silylated derivatives of polar compounds based on single ion Catharanthus roseus, Ocimum sanctum and Lantana camara on fecundity and
monitoring GC-MS. Int. J. Environ. Res. Publ. Health 16, 1–22. fertility of red cotton bug, Dysdercus koenigii Fabricius (heteroptera:
Guo, X.R., Yang, L., Yu, J.H., Tang, Z.H., Zu, Y.G., 2007. Alkaloid variations in pyrrhocoreidae). J. Entomol. Zool. Stud 5, 348–353.
Catharanthus roseus seedlings treated by different temperatures in short term and Kessler, M., Ubeaud, G., Jung, L., 2003. Anti-and pro-oxidant activity of rutin and
long term. J. For. Res. 18, 313–315. quercetin derivatives. J. Pharm. Pharmacol. 55, 131–142.
Gupta, M.M., Singh, D.V., Tripathi, A.K., Pandey, R., Verma, R.K., Singh, S., Shasany, A. Kevin, L.Y.W., Hussin, A.H., Zhari, I., Chin, J.H., 2012. Sub–acute oral toxicity study of
K., Khanuja, S.P.S., 2005. Simultaneous determination of vincristine, vinblastine, methanol leaves extract of Catharanthus roseus in rats. J. Acute Dis 1, 38–41.
catharanthine, and vindoline in leaves of Catharanthus roseus by high-performance Khalil, A., 2012. Antimicrobial activity of ethanol leaf extracts of Catharanthus roseus
liquid chromatography. J. Chromatogr. Sci. 43, 450–453. from Saudi Arabia. In 2nd international conference on environment science and
Gupta, P., 2009. Antiestrogenic activity of petroleum ether extracts of the leaves of biotechnology (Vol. 48, No. 2, 6–11.
Catharanthus roseus (Vinca rosea) in female albino mice. Asian J. Exp. Sci. 23, Khanavi, M., Pourmoslemi, S., Farahanikia, B., Hadjiakhoondi, A., Ostad, S.N., 2010.
313–316. Cytotoxicity of Vinca minor. Pharm. Biol. 48, 96–100.
Gupta, S., Pandey-Rai, S., Srivastava, S., Naithani, S.C., Prasad, M., Kumar, S., 2007. King, K.L., Boder, G.B., 1979. Correlation of the clinical neurotoxicity of the vinca
Construction of genetic linkage map of the medicinal and ornamental plant alkaloids vincristine, vinblastine, and vindesine with their effects on cultured rat
Catharanthus roseus. J. Genet. 86, 259–268. midbrain cells. Cancer Chemother. Pharmacol 2, 239–242.
Gurib-Fakim, A., Sewraj, M.D., Gueho, J., Dulloo, E., 1996. Medicinal plants of Knobloch, K.H., Bast, G., Berlin, J., 1982. Medium-and light-induced formation of
Rodrigues. J. Pharmacogn. Phytochem. 34, 2–14. serpentine and anthocyanins in cell suspension cultures of Catharanthus roseus.
Hanafy, M.S., Matter, M.A., Asker, M.S., Rady, M.R., 2016. Production of indole alkaloids Phytochemistry 21, 591–594.
in hairy root cultures of Catharanthus roseus L. and their antimicrobial activity. S. Koel, M., Kuhtinskaja, M., Vaher, M., 2020. Extraction of bioactive compounds from
Afr. J. Bot., Le 105, 9–18. Catharanthus roseus and Vinca minor. Sep. Purif. Technol. 252, 117438.
Hashemabadi, D., Sabzevari, F., Kaviani, B., Ansari, M.H., 2018. Organic N-fertilizer, Konermann, 2017. Addressing a common misconception: ammonium acetate as neutral
rhizobacterial inoculation and fungal compost improve nutrient uptake, plant pH “buffer” for native electrospray mass spectrometry. J. Am. Soc. Mass Spectrom.
growth and the levels of vindoline, ajmalicine, vinblastine, catharanthine and total 28, 1827–1835.
alkaloids in Catharanthus roseus L. Folia Hortic 30, 203–213. Kratz, J.M., Andrighetti-Fröhner, C.R., Kolling, D.J., Leal, P.C., Cirne-Santos, C.C.,
Hassan, K.A., Brenda, A.T., Patrick, V., Patrick, O.E., 2011. In vivo antidiarrheal activity Yunes, R.A., Nunes, R.J., Trybala, E., Bergström, T., Frugulhetti, I.C., Barardi, C.R.
of the ethanolic leaf extract of Catharanthus roseus Linn. (Apocyanaceae) in Wistar M., 2008. Anti-HSV-1 and anti-HIV-1 activity of gallic acid and pentyl gallate. Mem.
rats. Afr. J. Pharm. Pharmaco 5, 1797–1800. Inst. Oswaldo Cruz 103, 437–442.
He, L., Yang, L., Xiong, A., Zhao, S., Wang, Z., Hu, Z., 2011. Simultaneous quantification Krieglstein, J., Rischke, R., 1991. Vinpocetine increases the neuroprotective effect of
of four indole alkaloids in Catharanthus roseus cell line C20hi by UPLC-MS. Anal. Sci. adenosine in vitro.Eur. J. Pharmacol 205, 7–10.
27, 433–433. Kris, M.G., Gralla, R.J., Kalman, L.A., Kelsen, D.P., Casper, E.S., Burke, M.T., Groshen, S.,
Hiruki, C., da Rocha, A., 1986. Histochemical diagnosis of mycoplasma infections in Cibas, I.R., Bagin, R., Heelan, R.T., 1985. Randomized trial comparing vindesine plus
Catharanthus roseus by means of a fluorescent DNA-binding agent, 4, 6-diamidino-2- cisplatin with vinblastine plus cisplatin in patients with non-small cell lung cancer,
phenylindole-2HCl (DAPI). Can. J. Plant Pathol. 8, 185–188. with an analysis of methods of response assessment. Cancer Treat. For. Rep. 69,
Hisiger, S., Jolicoeur, M., 2007. Analysis of Catharanthus roseus alkaloids by HPLC. 387–395.
Phytochemistry Rev. 6, 207–234. Kroes, B.V., Van den Berg, A.J.J., Van Ufford, H.Q., Van Dijk, H., Labadie, R.P., 1992.
Hoskeri, H., Agarwal, S., Jacob, S., Chettri, N., Bisoyi, S., Tazeen, A., Vedamurthy, A., Anti-inflammatory activity of gallic acid. Planta Med.58 499–504.
Krishna, V., 2011. In-vitro anthelminthic activity of Catharanthus roseus extract. Int. Kumar, A., Patil, D., Rajamohanan, P.R., Ahmad, A., 2013. Isolation, purification and
J. Pharmaceut. Sci. Drug Res. 3, 211–213. characterization of vinblastine and vincristine from endophytic fungus Fusarium
Inte, V.M.L., Ragasa, C.Y., Rideout, J.A., 1998. Triterpenes, hydrocarbons and an oxysporum isolated from Catharanthus roseus. PloS One 8, 71805.
antimutagenic alkaloid from Catharanthus roseus (Apocynaceae). Asia Life Sci. 7, Kumar, S., Bagchi, G.D., Darokar, M.P., 1997. Antibacterial activity observed in the seeds
11–21. of some coprophilous plants. IJP 35, 179–184.
Islam, M.A., Akhtar, M.A., Islam, M.R., Hossain, M.S., Alam, M.K., Wahed, M.I.I., Kumar, A.G., Edward, C., Rajan, K., 2011. Antifeedant and toxicity of root extract of
Rahman, B.M., Anisuzzaman, A.S.M., Shaheen, S.M., Ahmed, M., 2009a. Catharanthus roseus (L.) G. Don on the tobacco leaf armyworm, Spodoptera litura
Antidiabetic and hypolipidemic effects of different fractions of Catharanthus roseus (Fabricius). Int. J. Biores 2, 7–10.
(Linn.) on normal and streptozotocin-induced diabetic rats. J. Sci. Res. 1, 334–344. Kumar, S., Bajpai, V., Singh, A., Kumar, B., 2018a. Identification, characterization and
Islam, M.A., Akhtar, M.A., Khan, M.R., Hossain, M.S., Alam, A.H., Ibne-Wahed, M.I., distribution of terpene indole alkaloids in ethanolic extracts of Catharanthus roseus
Amran, M.S., Rahman, B.M., Ahmed, M., 2009b. Oral glucose tolerance test (OGTT) using high-performance liquid chromatography/electrospray ionization quadrupole
in normal control and glucose induced hyperglycemic rats with Coccinia cordifolia L. time-of-flight tandem mass spectrometry and the study of their geographical
and Catharanthus roseus L. Pak. J. Pharm. Sci. 22, 402–404. variation. Rapid Commun. Mass Spectrom. 32, 319–332.
Iweala, E.E., Okeke, C.U., 2005. Comparative study of the hypoglycemic and biochemical Kumar, S., Singh, A., Bajpai, V., Srivastava, M., Singh, B.P., Kumar, B., 2016a. Structural
effects of Catharanthus roseus (Linn) g. apocynaceae (Madagascar periwinkle) and characterization of monoterpene indole alkaloids in ethanolic extracts of Rauwolfia
chlorpropamide (diabenese) on alloxan-induced diabetic rats. Biokemistri 17, species by liquid chromatography with quadrupole time-of-flight mass spectrometry.
149–156. J. Pharm. Anal 6, 363–373.
Jacobs, D.I., Snoeijer, W., Hallard, D., Verpoorte, R., 2004. The Catharanthus alkaloids: Kumar, S., Singh, A., Bajpai, V., Srivastava, M., Singh, B.P., Ojha, S., Kumar, B., 2016b.
pharmacognosy and biotechnology. Curr. Med. Chem. 11, 607–628. Simultaneous determination of bioactive monoterpene indole alkaloids in ethanolic
James, S.A., Bilbiss, L., Muhammad, B.Y., 2007. The effects of Catharanthus roseus (L) G. extract of seven Rauvolfia species using UHPLC with hybrid triple quadrupole linear
Don 1838 aqueous leaf extract on some liver enzymes, serum proteins and vital ion trap mass spectrometry. Phytochem. Anal. 27, 296–303.
organs. Sci. World J. 2, 5–9. Kumar, S., Singh, A., Kumar, B., Singh, B., Bahadur, L., Lal, M., 2018b. Simultaneous
Jasmine, D.J., Agastian, P., 2013. In vitro antioxidant activity and in vivo alpha quantitative determination of bioactive terpene indole alkaloids in ethanolic extracts
glucosidase activity of endophytic actinomycetes isolated from Catharanthus roseus of Catharanthus roseus (L.) G. Don by ultra-high performance liquid
(l.) G. Don. J. Pharm. Res. 6, 674–678. chromatography–tandem mass spectrometry. J. Pharmaceut. Biomed. Anal. 151,
Jayanthi, M., Sowbala, N., Rajalakshmi, G., Kanagavalli, U., Sivakumar, V., 2010. Study 32–41.
of antihyperglycemic effect of Catharanthus roseus in alloxan induced diabetic rats. Kumari, K., Gupta, S., 2013. Antifungal properties of leaf extract of Catharanthus roseus l
Int. J Pharm. Pharmaceut. Sci. 2, 114–116. (g.) Don. AJPCT 1, 698–705.
Jeong, W.T., Lim, H.B., 2018. A UPLC-ESI-Q-TOF method for rapid and reliable Kuppusamy, C., Murugan, K., Arul, N., Yasodha, P., 2009. Larvicidal and insect growth
identification and quantification of major indole alkaloids in Catharanthus roseus. regulator effect of α-amyrin acetate from Catharanthus roseus Linn against the
J. Chromatogr. B 1080, 27–36. malaria vector Anopheles Stephensi Liston (Diptera: Culicidae). J. Entomol. Res. 39,
Junaid, A., Mujib, A., Sharma, M.P., Tang, W., 2007. Growth regulators affect primary 78–83.
and secondary somatic embryogenesis in Madagaskar periwinkle (Catharanthus Lahare, R.P., Yadav, H.S., Dashhare, A., Bisen, Y.K., 2020. An updated review on
roseus (L.) G. Don) at morphological and biochemical levels. Plant Growth Regul. 51, phytochemical and pharmacological properties of Catharanthus rosea. Saudi J. Med.
271–281. Pharm. Sci. 6, 759–766.
Kabesh, K., Senthilkumar, P., Ragunathan, R., Kumar, R.R., 2015. Phytochemical Lakics, V., Sebestyén, M.G., Erdö, S.L., 1995. Vinpocetine is a highly potent
analysis of Catharanthus roseus plant extract and its antimicrobial activity. Int. J. neuroprotectant against veratridine-induced cell death in primary cultures of rat
Pure App. Bios 3, 162–172. cerebral cortex. Neurosci. Lett. 185, 127–130.
Kabubii, Z.N., Mbaria, J.M., Mbaabu, M., 2015. Acute toxicity studies of Catharanthus Lamar, J.C., Poignet, H., Beaughard, M., Dureng, G., 1988. Calcium antagonist activity of
roseus aqueous extract in male Wistar rats. Afr. J. Pharmacol. Ther 4, 130–134. vinpocetine and vincamine in several models of cerebral ischaemia. Drug Dev. Res.
Kalidass, C., Mohan, V.R., Daniel, A., 2009. Effect of auxin and cytokinin on vincristine 14, 297–304.
production by callus cultures of Catharanthus roseus L. (apocynaceae). Trop. Subtrop. Lawal, O.A., Ogunwande, I.A., Ibirogba, A.E., Layode, O.M., Opoku, A.R., 2015.
Agroecosyst 12, 283–288. Chemical constituents of essential oils from Catharanthus roseus (L.) G. Don grown in
Kaplan, L., Abrams, D., Volberding, P., 1986. Treatment of Kaposi’s sarcoma in acquired Nigeria. J. Essent 18, 57–63.
immunodeficiency syndrome with an alternating vincristine-vinblastine regimen. Lee, C.W., Shuler, M.L., 2000. The effect of inoculum density and conditioned medium
Cancer Treat. For. Rep. 70, 1121–1122. on the production of ajmalicine and catharanthine from immobilized Catharanthus
Karimi, M., Raofie, F., 2019. Micronization of vincristine extracted from Catharanthus roseus cells. Biotechnol. Bioeng 67, 61–71.
roseus by expansion of supercritical fluid solution. J. Supercrit. Fluids 146, 172–179.

40
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Lee-Parsons, C.W., Ertürk, S., Tengtrakool, J., 2004. Enhancement of ajmalicine Mu, F., Yang, L., Wang, W., Luo, M., Fu, Y., Guo, X., Zu, Y., 2012. Negative-pressure
production in Catharanthus roseus cell cultures with methyl jasmonate is dependent cavitation extraction of four main vinca alkaloids from Catharanthus roseus leaves.
on timing and dosage of elicitation. Biotechnol. Lett. 26, 1595–1599. Molecules 17, 8742–8752.
Levac, D., Murata, J., Kim, W.S., De Luca, V., 2008. Application of carborundum abrasion Muralidharan, L., 2014. Catharanthus roseus Leaves as an anti-diabetic and hypolipidemic
for investigating the leaf epidermis: molecular cloning of Catharanthus roseus 16- agents in alloxan-induced diabetic rats. AJPCT 2, 1393–1396.
hydroxytabersonine-16-O-methyltransferase. Plant J. 53, 225–236. Murata, J., Luca, V.D., 2005. Localization of tabersonine 16-hydroxylase and 16-OH
Levêque, D., Jehl, F., 2007. Molecular pharmacokinetics of catharanthus (vinca) tabersonine-16-O-methyltransferase to leaf epidermal cells defines them as a major
alkaloids. J. Clin. Pharmacol. 47, 579–588. site of precursor biosynthesis in the vindoline pathway in Catharanthus roseus. Plant
Li, G.Z., Hu, Y.H., Li, D.Y., Zhang, Y., Guo, H.L., Li, Y.M., Chen, F., Xu, J., 2020. J. 44, 581–594.
Vincristine-induced peripheral neuropathy: a mini-review. Neurotoxicology 81, Mustafa, N.R., Verpoorte, R., 2007. Phenolic compounds in Catharanthus roseus.
161–171. Phytochemistry Rev. 6, 243–258.
Li, M., Peebles, C.A., Shanks, J.V., San, K.Y., 2011. Effect of sodium nitroprusside on Naaranlahti, T., Nordström, M., Huhtikangas, A., Lounasmaa, M., 1987. Determination of
growth and terpenoid indole alkaloid production in Catharanthus roseus hairy root Catharanthus alkaloids by reversed-phase high-performance liquid chromatography.
cultures. Biotechnol. Prog. 27, 625–630. J. Chromatogr. A 410, 488–493.
Lin, C., Cai, J., Yang, X., Hu, L., Lin, G., 2015. Liquid chromatography mass spectrometry Naaranlahti, T., Ranta, V.P., Jarho, P., Nordström, M., Lapinjoki, S.P., 1989.
simultaneous determination of vindoline and catharanthine in rat plasma and its Electrochemical detection of indole alkaloids of Catharanthus roseus in high-
application to a pharmacokinetic study. Biomed. Chromatogr. 29, 97–102. performance liquid chromatography. Analyst 114, 1229–1231.
Liscombe, D.K., O’Connor, S.E., 2011. A virus-induced gene silencing approach to Naeem, M., Ansari, A.A., Aftab, T., Alam, M.M., Masroor, M., Khan, A., Uddin, M., 2019.
understanding alkaloid metabolism in Catharanthus roseus. Phytochemistry 72, Triacontanol improves production of anticancer alkaloids in Catharanthus roseus L.
1969–1977. AJPCR 11, 21–27.
Liu, D.H., Jin, H.B., Chen, Y.H., Cui, L.J., Ren, W.W., Gong, Y.F., Tang, K.X., 2007. Nagy-Turak, A., Vegh, Z., 1994. Extraction and in situ densitometric determination of
Terpenoid indole alkaloids biosynthesis and metabolic engineering in Catharanthus alkaloids from Catharanthus roseus by means of overpressured layer chromatography
roseus. J. Integr 49, 961–974. on amino-bonded silica layers I. Optimization and validation of the separation
Liu, Z., Wu, H.L., Li, Y., Gu, H.W., Yin, X.L., Xie, L.X., Yu, R.Q., 2016. Rapid and system. J. Chromatogr. A 668, 501–507.
simultaneous determination of five vinca alkaloids in Catharanthus roseus and human Nakabayashi, R., Mori, T., Takeda, N., Toyooka, K., Sudo, H., Tsugawa, H., Saito, K.,
serum using trilinear component modeling of liquid chromatography–diode array 2020. Metabolomics with 15N labeling for characterizing missing monoterpene
detection data. J. Chromatogr. B 1026, 114–123. indole alkaloids in plants. Anal. Chem. 92, 5670–5675.
Luo, M., Yang, L.Q., Yao, X.H., Mu, F.S., Zhang, D.Y., Song, Z.Y., Qiao, Q., Fu, Y.J., Zu, Y. Nakabayashi, R., Tsugawa, H., Kitajima, M., Takayama, H., Saito, K., 2015. Boosting
G., 2014. Optimization of enzyme-assisted negative pressure cavitation extraction of sensitivity in liquid chromatography–Fourier transform ion cyclotron
five main indole alkaloids from Catharanthus roseus leaves and its pilot-scale resonance–tandem mass spectrometry for product ion analysis of monoterpene
application. Sep. Purif. Technol. 125, 66–73. indole alkaloids. Front. Plant Sci. 6 https://doi.org/10.3389/fpls.2015.01127, 1127.
Magnotta, M., Murata, J., Chen, J., De Luca, V., 2007. Expression of deacetylvindoline-4- Nammi, S., Boini, M.K., Lodagala, S.D., Behara, R.B.S., 2003. The juice of fresh leaves of
O-acetyltransferase in Catharanthus roseus hairy roots. Phytochemistry 68, Catharanthus roseus Linn. reduces blood glucose in normal and alloxan diabetic
1922–1931. rabbits. BMC Complement. Altern. Med. Times 3, 1–4.
Malathi, V., Devi, S.S., Revathi, K., 2010. Anti diabetic activity by the in vitro alpha Nayak, B.S., Anderson, M., Pereira, L.P., 2007. Evaluation of wound-healing potential of
amylase and alpha-glucosidase inhibitory activity of Catharanthus roseus. Bioscan 5, Catharanthus roseus leaf extract in rats. Fitoterapia 78, 540–544.
655–659. Naz, S., Haq, R., Aslam, F., Ilyas, S., 2015. Evaluation of antimicrobial activity of extracts
Manda, V., Avula, B., Dale, O., Chittiboyina, A., Khan, I., Walker, L., Khan, S., 2015. of in vivo and in vitro grown Vinca rosea L. (Catharanthus roseus) against pathogens.
Studies on pharmacokinetic drug interaction potential of vinpocetine. Medicines 2, Pak. J. Pharm. Sci. 28, 849–853.
93–105. Nejat, N., Valdiani, A., Cahill, D., Tan, Y.H., Maziah, M., Abiri, R., 2015. Ornamental
Manoharan, S., Umadevi, S., Jayanthi, S., Baskaran, N., 2011. Antihyperglycemic effect exterior versus therapeutic interior of Madagascar periwinkle (Catharanthus roseus):
of Coscinium fenestratum and Catharanthus roseus in alloxan-induced diabetic rats. the two faces of a versatile herb. Sci. World J 2015, 1–19.
Int. J. Nutr. Pharmacol. Neurol Dis 1, 189–193. Nisar, A., 2017a. Antioxidant and total phenolic content of Catharanthus roseus using
Marcone, C., Ragozzino, A., Seemuller, E., 1997. Dodder transmission of alder yellows deep eutectic solvent. Recent Adv. Biol. Med 3, 7–10.
phytoplasma to the experimental host Catharanthus roseus (periwinkle). Eur. J. Plant Nisar, A., 2017b. Identification of flavonoids (quercetin, gallic acid and rutin) from
Pathol. 27, 347–350. Catharanthus roseus plant parts using deep eutectic solvent. Recent Adv. Biol. Med 3,
Mardani-Nejad, S., Khavari-Nejad, R.A., Saadatmand, S., Najafi, F., Azar, P.A., 2016. 1–6.
Potent antioxidant properties of rolB-transformed Catharanthus roseus (L.) G.don. Nisar, A., Mamat, A.S., Hatim, M.I., Aslam, M.S., Syarhabil, M., 2016. An updated review
Iran. J. Pharm 15, 537–550. on Catharanthus roseus: phytochemical and pharmacological analysis. IRJPS 9,
McDaniel, R., Weiss, R., 2005. Advances in synthetic biology: on the path from 631–653.
prototypes to applications. Curr. Opin. Biotechnol. 16, 476–483. Nishibe, S., Takenaka, T., Fujikawa, T., Yasukawa, K., Takido, M., Morimitsu, Y.,
Mehta, J., Upadhyay, D., Paras, P., Ansari, R., Rathore, S., Tiwari, S., 2013. Multiple Hirota, A., Kawamura, T., Noro, Y., 1996. Bioactivephenolic compounds from
shoots regeneration of (anticancer plant) Catharanthus roseus–an important Catharanthus roseus and Vinca minor. Nat. Med. 50, 378–383.
medicinal plant. Am. J. PharmTech Res. 3, 785–793. O’Connor, S.E., 2012. Strategies for engineering plant natural products: the iridoid-
Memelink, J., Gantet, P., 2007. Transcription factors involved in terpenoid indole derived monoterpene indole alkaloids of Catharanthus roseus. Methods in
alkaloid biosynthesis in Catharanthus roseus. Phytochem. Rev.6 353–362. Enzymology, vol. 515. Academic Press, Cambridge, Massachusetts, United States,
Mishra, J.N., Verma, N.K., 2017. A brief study on Catharanthus roseus: a review. Intern J. pp. 189–206.
Res. Pharmacy Pharmaceut Sci. 2, 20–23. O’Connor, S.E., Maresh, J.J., 2006. Chemistry and biology of monoterpene indole
Mishra, P., Uniyal, G.C., Sharma, S., Kumar, S., 2001. Pattern of diversity for alkaloid biosynthesis. Nat. Prod. Rep. 23, 532–547.
morphological and alkaloid yield related traits among the periwinkle Catharanthus Ohadoma, S.C., Michael, H.U., 2011. Effects of co—administration of methanol leaf
roseus accessions collected from in and around Indian subcontinent. Genet. Resour. extract of Catharanthus roseus on the hypoglycemic activity of metformin and
Crop. Ev 48, 273–286. glibenclamide in rats. Asian Pac. J. Trop. Med. 4, 475–477.
Mohan, S.C.T., Priyadarshini, G.S., Balamurugan, V., 2015. GC-MS analysis of Orviský, E., Šoltés, L., Stančıková, M., 1997. High-molecular-weight hyaluronan—a
phytochemicals and hypoglycemic effect of Catharathus roseus in alloxan induced valuable tool in testing the antioxidative activity of amphiphilic drugs stobadine and
diabetic rats. Int. J. Pharma Sci. Res. 25, 123–128. vinpocetine. J. Pharmaceut. Biomed. Anal. 16, 419–424.
Mahroug, S., Burlat, V., St-Pierre, B., 2007. Cellular and sub-cellular organisation of the Orwa, J.A., Mwitari, P.G., Matu, E.N., Rukunga, G.M., 2007. Traditional healers and the
monoterpenoid indole alkaloid pathway in Catharanthus roseus. Phytochemistry Rev. managment of malaria in kisumu district, Kenya. East afr. Med. J. 84, 51–55.
6, 363–381. Osterlind, K., Hørbov, S., Dombernowsky, P., Rørth, M., Hansen, H.H., 1982. Vindesine
Monforte-González, M., Ayora-Talavera, T., Maldonado-Mendoza, I.E., Loyola-Vargas, V. in the treatment of squamous cell carcinoma, adenocarcinoma, and large cell
M., 1992. Quantitative analysis of serpentine and ajmalicine in plant tissues of carcinoma of the lung. Cancer Treat. For. Rep. 66, 305–309.
Catharanthus roseus and hyoscyamine and scopolamine in root tissues of Datura Oudin, A., Courtois, M., Rideau, M., Clastre, M., 2007. The iridoid pathway in
stramonium by thin layer chromatography-densitometry. Phytochem. Anal. 3, Catharanthus roseus alkaloid biosynthesis. Phytochem.Rev. 6, 259–276.
117–121. Paarakh, M.P., Swathi, S., Taj, T., Tejashwini, V.T., Tejashwini, B., 2019. Catharanthus
Moon, S.H., Pandurangan, M., Kim, D.H., Venkatesh, J., Patel, R.V., Mistry, B.M., 2018. roseus L. -A review. Acta sci. Pharmaceut. Sci. 3, 19–24.
A rich source of potential bioactive compounds with anticancer activities by Palaniappan, N., Cole, I., Caballero-Briones, F., Manickam, S., Thomas, K.J., Santos, D.,
Catharanthus roseus cambium meristematic stem cell cultures. J. Ethnopharmacol. 2020. Experimental and DFT studies on the ultrasonic energy-assisted extraction of
217, 107–117. the phytochemicals of Catharanthus roseus as green corrosion inhibitors for mild steel
Moreno-Valenzuela, O.A., Galaz-Avalos, R.M., Minero-García, Y., Loyola-Vargas, V.M., in NaCl medium. RSC Adv. 10, 5399–5411.
1998. Effect of differentiation on the regulation of indole alkaloid production in Pan, Q., Chen, Y.U., Wang, Q., Yuan, F., Xing, S., Tian, Y., Zhao, J., Sun, X., Tang, K.,
Catharanthus roseus hairy roots. Plant Cell Rep. 18, 99–104. 2010. Effect of plant growth regulators on the biosynthesis of vinblastine, vindoline
Mostofa, M., Choudhury, M.E., Hossain, M.A., Islam, M.Z., Islam, M.S., Sumon, M.H., and catharanthine in Catharanthus roseus. Plant Growth Regul. 60, 133–141.
2007. Antidiabetic effects of Catharanthus roseus, Azadirachta indica, Allium Pan, Q., Mustafa, N.R., Tang, K., Choi, Y.H., Verpoorte, R., 2016. Monoterpenoid indole
sativum and glimepride in experimentally diabetic induced rat. J. Adv. Vet. Anim. alkaloids biosynthesis and its regulation in Catharanthus roseus: a literature review
Med 5, 99–102. from genes to metabolites. Phytochem. Rev.15 221–250.
Pandey, S.S., Singh, S., Babu, C.V., Shanker, K., Srivastava, N.K., Shukla, A.K., Kalra, A.,
2016. Fungal endophytes of Catharanthus roseus enhance vindoline content by

41
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

modulating structural and regulatory genes related to terpenoid indole alkaloid Ramya, S., 2008. In vitro evaluation of antibacterial activity using crude extracts of
biosynthesis. Sci. Rep. 6, 1–14. Catharanthus roseus L. (G.) Don. Ethnobot. Leaf 12, 1076–1072.
Pandey-Rai, S., Mallavarapu, G.R., Naqvi, A.A., Yadav, A., Rai, S.K., Srivastava, S., Rao, A.S., Ahmed, M.F., 2014. Simultaneous determination of phenolic compounds in
Singh, D., Mishra, R., Kumar, S., 2006. Volatile components of leaves and flowers of Catharanthus roseus leaves by HPLC method. Int. J. Pharm. Sci. 5, 977–981.
periwinkle Catharanthus roseus (L.) G. Don from New Delhi. Flavour Fragrance J. 21, Rasineni, K., Bellamkonda, R., Singareddy, S.R., Desireddy, S., 2010. Antihyperglycemic
427–430. activity of Catharanthus roseus leaf powder in streptozotocin-induced diabetic rats.
Pandiangan, D., Tilaar, W., Nainggolan, N., Wahyudi, L., 2013. Relations between Phcog. Mag. 2, 195–201.
catharanthine content enhancement with the other associated secondary metabolites Rasool, N., Rizwan, K., Zubair, M., Naveed, K.U.R., Imran, I., Ahmed, V.U., 2011.
in Catharanthus roseus cell culture that treated tryptophan. Int. J. Sci. Res. 4, Antioxidant potential of different extracts and fractions of Catharanthus roseus
2208–2212. shoots. Int. J. Phytomed. 3, 108–114.
Panneerselvam, C., Murugan, K., Kovendan, K., Kumar, P.M., Ponarulselvam, S., Raza, M.L., Nasir, M., Abbas, T., Naqvi, B.S., 2009. Antibacterial activity of different
Amerasan, D., Subramaniam, J., Hwang, J.S., 2013. Larvicidal efficacy of extracts from the Catharanthus roseus. Clin. Exp. Med. 3, 81–85.
Catharanthus roseus Linn. (Family: apocynaceae) leaf extract and bacterial insecticide Ren, T., Li, R., Meng, X., Fawcett, J.P., Sun, D., Gu, J., 2018. Differential mobility
Bacillus thuringiensis against Anopheles stephensi Liston. Asian Pac. J. Trop. Med. 6, spectrometry followed by tandem mass spectrometry with multiple ion monitoring
847–853. for bioanalysis of eptifibatide in rat plasma. J. Pharmaceut. Biomed 151, 260–265.
Parthasarathy, R., Shanmuganathan, R., Pugazhendhi, A., 2020. Vinblastine production Renaudin, J.P., 1984. Reversed-phase high-performance liquid chromatographic
by the endophytic fungus Curvularia verruculosa from the leaves of Catharanthus characteristics of indole alkaloids from cell suspension cultures of Catharanthus
roseus and its in vitro cytotoxicity against HeLa cell line. Anal. Biochem. 593, 113530 roseus. J. Chromatogr. A 291, 165–174.
https://doi.org/10.1016/j.ab.2019.113530. Renaudin, J.P., 1985. Extraction and fluorimetric detection after high-performance
Patel, Y., Vadgama, Vishalkuma, Baxi, S., Tripathi, C.B., 2011. Evaluation of liquid chromatography of indole alkaloids from cultured cells of Catharanthus
hypolipidemic activity of leaf juice of Catharanthus roseus (Linn.) G. Donn. in Guinea roseus. Physiol. Veg. 23, 381–388.
pigs. Acta Pol. Pharm. Times 68, 927–935. Renjini, K.R., Gopakumar, G., Latha, M.S., 2017. The medicinal properties of
Patil, P.J., Ghosh, J.S., 2010. Antimicrobial activity of Catharanthus roseus–a detailed phytochemicals in catharanthus roseus-A review. Eur. J. Pharm. Med. Res.4
study. Br. J. Pharmacol. 1, 40–44. 545–551.
Pavunraj, M., Ramesh, V., Sakthivelkumar, S., Veeramani, V., Janarthanan, S., 2017. Retna, M.A., Ethalsa, P., 2013. A review of the taxonomy, ethnobotany, chemistry and
Larvicidal and enzyme inhibitory effects of acalypha fruticosa (F.) and Catharanthus pharmacology of Catharanthus roseus (Apocyanaceae). Int. J. Res. Eng. Technol 2,
roseus L (G) don. Leaf extracts against Culex quinquefasciatus (say.)(Diptera: 3899–3912.
Culicidae). Asian J. Pharm. Clin. Res.10 213–220. Rijhwani, S.K., Shanks, J.V., 1998. Effect of elicitor dosage and exposure time on
Peebles, C.A., Hong, S.B., Gibson, S.I., Shanks, J.V., San, K.Y., 2006. Effects of terpenoid biosynthesis of indole alkaloids by Catharanthus roseus hairy root cultures.
precursor feeding on Catharanthus roseus hairy roots over-expressing the alpha or the Biotechnol. Prog. 14, 442–449.
alpha and beta subunits of anthranilate synthase. Biotechnol. Bioeng. 93, 534–540. Rischer, H., Orešič, M., Seppänen-Laakso, T., Katajamaa, M., Lammertyn, F., Ardiles-
Pennanen, S., Huhtikangas, A., 1990. Photochemical one-pot synthesis of vinblastine and Diaz, W., Van Montagu, M.C., Inzé, D., Oksman-Caldentey, K.M., Goossens, A., 2006.
vincristine. J. Photochem. Photobiol., A 51, 515–518. Gene-to-metabolite networks for terpenoid indole alkaloid biosynthesis in
Pereira, D.M., Faria, J., Gaspar, L., Ferreres, F., Valentão, P., Sottomayor, M., Andrade, P. Catharanthus roseus cells. Proc. Natl. Acad. Sci. Unit. States Am. 103, 5614–5619.
B., 2010. Exploiting Catharanthus roseus roots: source of antioxidants. Food Chem. Rosenthal, S., Kaufman, S., 1974. Vincristine neurotoxicity. Ann. Intern. Med. 80,
121, 56–61. 733–737.
Pham, H.N.T., Sakoff, J.A., Van Vuong, Q., Bowyer, M.C., Scarlett, C.J., 2018. Screening Ross, I.A., 2003. Catharanthus Roseus. In: Medicinal Plants of the World (175-195).
phytochemical content, antioxidant, antimicrobial and cytotoxic activities of Humana Press, Totowa, NJ.
Catharanthus roseus (L.) G. Don stem extract and its fractions. Biocatal. Agric. Roy, S., Chatterjee, P., 2010. A non-toxic antifungal compound from the leaves of
Biotechnol 16, 405–411. Catharanthus roseus characterized as 5-hydroxy flavone by UV spectroscopic analysis
Pham, H.N.T., Vuong, Q.V., Bowyer, M.C., Scarlett, C.J., 2017. Effect of extraction and evaluation of its antifungal property by agar-cup method. Ind. Crop Prod. 32,
solvents and thermal drying methods on bioactive compounds and antioxidant 375–380.
properties of Catharanthus roseus (L.) G. Don (Patricia White cultivar). J. Food Ruszkowska, J., Chrobak, R., Wróbel, J.T., Czarnocki, Z., 2003. Novel Bisindole
Process. Pres 41, 1–8. Derivatives of Catharanthus Alkaloids with Potential Cytotoxic Properties. In:
Pietrosiuk, A., Furmanowa, M., Łata, B., 2007. Catharanthus roseus: micropropagation Developments in Tryptophan and Serotonin Metabolism. Springer, Boston, MA,
and in vitro techniques. Phytochem. Rev.6 459–473. pp. 643–646.
Piovan, A., Filippini, R., 2007. Anthocyanins in Catharanthus roseus in vivo and in vitro: a Sadique, A.S., Hashim, K.M., 2019. Anti-angiogenesis studies of the aerial parts of
review. Phytochemistry Rev. 6, 235–242. Catharanthus roseus with special emphasis to ethyl acetate extraction. Int. J. Innov.
Piovan, A., Filippini, R., Favretto, D., 1998. Characterization of the anthocyanins of Res. Sci. Eng. Technol.4 376–378.
Catharanthus roseus (L.) G. Don in vivo and in vitro by electrospray ionization ion Sagar, P.K., Meena, R.P., Ahmad, M.W., Sajwan, K., 2020. 2020. Useful anti-cancerous &
trap mass spectrometry. Rapid Commun. Mass Spectrom. 12, 361–367. anti-tumorous asian medicinal plants (Taxus baccata L. or Taxus baccata Thunb.,
Ponarulselvam, S., Panneerselvam, C., Murugan, K., Aarthi, N., Kalimuthu, K., Catharanthus roseus (L.) G. Don, Annona muricata L.). SIJTCM 5, 1–12.
Thangamani, S., 2012. Synthesis of silver nanoparticles using leaves of Catharanthus Sandey, J., Summarwar, S., 2016. Pupicidal activity of plant extracts of Catharanthus
roseus Linn. G. Don and their antiplasmodial activities. Asian Pac. J. Trop. Biomed 2, roseus against 6th instar of Spodoptera litura.Int. J. Fauna. Biol. Stud 3, 9–10.
574–580. Saravanan, S., Karthi, S., 2014. HPLC analysis for methanolic extract of Catharanthus
Prasad, S.K., Kulshreshtha, A., Qureshi, T.N., 2009. Antidiabetic activity of some herbal roseus under elevated CO2. World J. Pharmaceut. Sci. 3, 683–693.
plants in streptozotocin induced diabetic albino rats. Pakistan J. Nutr. 8, 551–557. Sathiya, S., Karthikeyan, B., Jaleel, A.B., Azooz, M.M., Iqbal, M., 2008. Antibiogram of
Quan, P.M., Binh, V.N., Ngan, V.T., Trung, N.T., Anh, N.Q., 2019. Molecular docking Catharanthus roseus extracts. Global J. Mol. Sci. 3, 1–7.
studies of Vinca alkaloid derivatives on Tubulin. Vietnam J. Sci. Technol. 57, Schmidt, M.S., Huang, R., Classon, R.J., Murry, D.J., 2006. Determination of vincristine
702–706. in infant plasma by liquid chromatography-atmospheric pressure chemical
Rahim, R.A., Ahmad, N.H., Al Azzam, K.M., Mat, I., 2018. Determination and ionization-mass spectroscopy. J. Pharm. Biomed 41, 540–543.
quantification of the vinblastine content in purple, red, and white Catharanthus Schröder, G., Unterbusch, E., Kaltenbach, M., Schmidt, J., Strack, D., De Luca, V.,
roseus leaves using RP-HPLC method. Adv. Pharmaceut. Bull. 8, 157–161. Schröder, J., 1999. Light-induced cytochrome P450-dependent enzyme in indole
Rahman, M.A., Mossa, J.S., Al-Said, M.S., Al-Yahya, M.A., 2004. Medicinal plant alkaloid biosynthesis: tabersonine 16-hydroxylase. FEBS Lett. 458, 97–102.
diversity in the flora of Saudi Arabia 1: a report on seven plant families. Fitoterapia Senbagalakshmi, P., Rao, M.V., Kumar, T.S., 2017. In vitro studies, biosynthesis of
75, 149–161. secondary metabolites and pharmacological utility of Catharanthus roseus (L.). In:
Rai, V., Tandon, P.K., Khatoon, S., 2014. Effect of chromium on antioxidant potential of G. Don.: a review. Catharanthus roseus. Springer, Cham, pp. 153–199.
Catharanthus roseus varieties and production of their anticancer alkaloids: vincristine Shams, K., Nazif, N., Azim, N., Shafeek, K., Missiry, M., Ismail, S., Nasr, M., 2009.
and vinblastine. BioMed Res. Int. 2014, 934182 https://doi.org/10.1155/2014/ Isolation and characterization of antineoplastic alkaloids from Catharanthus roseus l.
934182. Don. cultivated in Egypt. Afr. J. Tradit., Complementary Altern. Med. 6, 118–122.
Rajagopal, T., Jemimah, I.A.A., Ponmanickam, P., Ayyanar, M., 2015. Synthesis of silver Shanmugaraju, V., Bhakyaraj, R., 2016. Antimicrobial potential activity of leaf extracts
nanoparticles using Catharanthus roseus root extract and its larvicidal effects. of Catharanthus roseus against human pathogens under laboratory conditions. Int. J.
J. Environ. Biol. 36, 1283–1289. Curr. Res. Biol. Med 1, 35–51.
Ramani, S., Jayabaskaran, C., 2008. Enhanced catharanthine and vindoline production in Shaw, K., Nandi, K., Ganguly, R., Roy, C., 2017. Role of Catharanthus roseus Linn.
suspension cultures of Catharanthus roseus by ultraviolet-B light. J. Mol. Signal. 3, 9. (Nayantara) on sperm count and serum testosterone level of adult male albino rat.
https://doi.org/10.1186/1750-2187-3-9. World J. Pharm. Res. (N. Y.) 6, 1087–1096.
Rambhai, P.A., Sisodia, S.S., Chaudhuri, A., Patidar, A., 2019. Antiulcer and Shukla, A.K., Shasany, A.K., Verma, R.K., Gupta, M.M., Mathur, A.K., Khanuja, S.P.,
antihyperlipidaemic efficacy of Catharanthus roseus leaves on pylorus ligation 2010. Influence of cellular differentiation and elicitation on intermediate and late
induced ulcer in experimental animals. Pharmeuropa Bio J. 9, 7–12. steps of terpenoid indole alkaloid biosynthesis in Catharanthus roseus. Protoplasma
Ramirez, A., García-Rubio, S., 2003. Current progress in the chemistry and pharmacology 242, 35–47.
of akuammiline alkaloids. Curr. Med. Chem. 10, 1891–1915. Siddiqui, M.J., Ismail, Z., Aisha, A.F.A., Majid, A.M., 2010. Cytotoxic activity of
Ramos, A.E.F., Le Pogam, P., Alcover, C.F., N’Nang, E.O., Cauchie, G., Hazni, H., Catharanthus roseus (Apocynaceae) crude extracts and pure compounds against
Awang, K., Bréard, D., Echavarren, A.M., Frederich, M., Gaslonde, T., 2019. human colorectal carcinoma cell line. Int. J. Pharmacol. 6, 43–47.
Collected mass spectrometry data on monoterpene indole alkaloids from natural Siddiqui, M.J.A., Ismail, Z., Saidan, N.H., 2011. Simultaneous determination of
product chemistry research. Sci. Data 6, 1–6. secondary metabolites from Vinca rosea plant extractives by reverse phase high
performance liquid chromatography. Phcog. Mag. 7, 92–96.

42
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

Sim, S.J., Chang, H.N., Liu, J.R., Jung, K.H., 1994. Production and secretion of indole Tin-Wa, M., Fong, H.H.S., Blomster, R.N., Farnsworth, N.R., 1968. Catharanthus
alkaloids in hairy root cultures of Catharanthus roseus: effects of in situ adsorption, alkaloids XXI. Isolation of lochnerinine, one of the cytotoxic principles of C. Pusillus.
fungal elicitation and permeabilization. J. Biosci. Bioeng. 78, 229–234. J. Pharm. Sci. 57, 2167–2169.
Simirgiotis, M.J., Silva, M., Becerra, J., Schmeda-Hirschmann, G., 2012. Direct Tiong, S., Looi, C., Hazni, H., Arya, A., Paydar, M., Wong, W., Cheah, S.C., Mustafa, M.,
characterisation of phenolic antioxidants in infusions from four Mapuche medicinal Awang, K., 2013. Antidiabetic and antioxidant properties of alkaloids from
plants by liquid chromatography with diode array detection (HPLC-DAD) and Catharanthus roseus (L.) G. Don. Molecules 18, 9770–9784.
electrospray ionisation tandem mass spectrometry (HPLC-ESI–MS). Food Chem. 131, Tiong, S.H., Looi, C.Y., Arya, A., Wong, W.F., Hazni, H., Mustafa, M.R., Awang, K., 2015.
318–327. Vindogentianine, a hypoglycemic alkaloid from Catharanthus roseus (L.) G. Don
Singh, A., Singh, P.K., Singh, R.K., 2014. Antidiabetic and wound healing activity of (Apocynaceae). Fitoterapia 102, 182–188.
Catharanthus roseus L. in streptozotocin-induced diabetic mice. Am. J. Phytomed. Toivonen, L., Ojala, M., Kauppinen, V., 1991. Studies on the optimization of growth and
Clin. Ther. 2, 686–692. indole alkaloid production by hairy root cultures of Catharanthus roseus. Biotechnol.
Singh, B., 2020. Botanical Leads for Drug Discovery. In: Springer Nature. Singapore Pte Bioeng. 37, 673–680.
Ltd., Singapore, p. 340. https://doi.org/10.1007/978-981-15-5917-4. Tong, L., Zhang, B., Tang, Z., Guo, X., Jia, X., Yu, J., 2011. Effects of low light stress on
Singh, D., Rai, S.K., Pandey-Rai, S., Srivastava, S., Mishra, R.K., Chaudhary, S., the biomass distribution and secondary metabolism of Catharanthus roseus. Adv.
Sharma, S., Kumar, S., 2008. Predominance of the serpentine route in Bot. Res. 31, 227–230.
monoterpenoid indole alkaloid pathway of Catharanthus roseus. In Proc Indian Natl Tsuruo, T., Iida, H., Tsukagoshi, S., Sakurai, Y., 1981. Overcoming of vincristine
Sci Acad 74 (3), 97–109. resistance in P388 leukemia in vivo and in vitro through enhanced cytotoxicity of
Singh, D.V., Maithy, A., Verma, R.K., Gupta, M.M., Kumar, S., 2000. Simultaneous vincristine and vinblastine by verapamil. Cancer Res. 41, 1967–1972.
determination of Catharanthus alkaloids using reversed phase high performance Uniyal, G.C., Bala, S., Mathur, A.K., Kulkarni, R.N., 2001. Symmetry C18 column: a
liquid chromatography. J. Liq. Chromatogr. Relat 20, 601–607. better choice for the analysis of indole alkaloids of Catharanthus roseus. Phytochem.
Singh, N., Kaushik, N.K., Mohanakrishnan, D., Tiwari, S.K., Sahal, D., 2015. Anal. 12, 206–210.
Antiplasmodial activity of medicinal plants from Chhotanagpur plateau, Jharkhand, Van der Heijden, R., Lamping, P.J., Out, P.P., Wijnsma, R., Verpoorte, R., 1987. High-
India.J. Ethnopharmacol 165, 152–162. performance liquid chromatographic determination of indole alkaloids in a
Singh, S.N., Vats, P., Suri, S., Shyam, R., Kumria, M.M.L., Ranganathan, S., Sridharan, K., suspension culture of Tabernaemontana divaricata. J. Chromatogr. A 396, 287–295.
2001. Effect of an antidiabetic extract of Catharanthus roseus on enzymic activities in Vardhan, S., Yadav, A.K., Suri, R.K., Nag, B.B.S.P., 2012. Genetic relatidness and in vitro
streptozotocin induced diabetic rats. J. Ethnopharmacol. 76, 269–277. antimicrobial activities of alkaloids isolated from Indian varieties of Catharanthus
Soares, P.R., de Oliveira, P.L., de Oliveira, C.M., Kato, L., Guillo, L.A., 2012. In vitro roseus (L.). Curr. Bot. 3, 43–48.
antiproliferative effects of the indole alkaloid vallesiachotamine on human Vega-Ávila, E., Cano-Velasco, J.L., Alarcón-Aguilar, F.J., Fajardo Ortíz, M.D.C., Almanza-
melanoma cells. Arch. Pharm. Res .35 565–571. Pérez, J.C., Román-Ramos, R., 2012. Hypoglycemic activity of aqueous extracts from
Srinivasan, D., Nathan, S., Suresh, T., Perumalsamy, P.L., 2001. Antimicrobial activity of Catharanthus roseus. Evid.-Based Complementary Altern. Med. Times 2012, 1–7.
certain Indian medicinal plants used in folkloric medicine. J. Ethnopharmacol. 74, Verma, A., Hartonen, K., Riekkola, M.L., 2008. Optimisation of supercritical fluid
217–220. extraction of indole alkaloids from Catharanthus roseus using experimental design
Steinmann, D., Ganzera, M., 2011. Recent advances on HPLC/MS in medicinal plant methodology—comparison with other extraction techniques. Phytochem. Anal 19,
analysis. J. Pharmaceut. Biomed. Anal. 55, 744–757. 52–63.
Suarsana, I.N., Utama, I.H., Kardena, I.M., Suartini, I.G.A.A., Watiniasih, N.L., 2015. Verma, A., Laakso, I., Seppänen-Laakso, T., Huhtikangas, A., Riekkola, M.L., 2007.
Hypolipidemic activity and low atherogenic index of aqueous leaf extracts of A simplified procedure for indole alkaloid extraction from Catharanthus roseus
Catharanthus roseus in hypercholesterolemic rats. J. Vet. 16, 533–541. combined with a semi-synthetic production process for vinblastine. Molecules 12,
Subarani, S., Sabhanayakam, S., Kamaraj, C., Elango, G., Kadir, M.A., 2013. Efficacy of 1307–1315.
larvicidal and pupicidal activity of Catharanthus roseus aqueous and solvent extracts Verma, P., Mathur, A.K., Arpan, S., Srivastava, A., Masood, N., Luqman, S.,
against Anopheles stephensi Liston and Culex quinquefasciatus Say (Diptera: Upadhyaya, M., Mathur, A., 2012. Co-culturing of Catharanthus roseus, Vinca major
Culicidae). Asian Pac. J. Trop. Med. 6, 625–630. and Rauwolfia serpentina cell suspensions in shake flask and bioreactor: production
Sukumar, K., Osmani, Z., 1981. Insect sterilants from Catharanthus roseus. Curr. Sci. 50, of a novel alkaloid with antioxidant potential. J. Med. Plants Res. 6, 4978–4988.
552–553. Verma, P., Mathur, A.K., Masood, N., Luqman, S., Shanker, K., 2013. Tryptophan over-
Susa, A.C., Xia, Z., Williams, E.R., 2017. Native mass spectrometry from common buffers producing cell suspensions of Catharanthus roseus (L) G. Don and their up-scaling in
with salts that mimic the extracellular environment. Angew. Chem. Int. Ed. 56, stirred tank bioreactor: detection of a phenolic compound with antioxidant
7912–7915. potential. Protoplasma 250, 371–380.
Syeda, A.M., Riazunnisa, K., 2020. Data on GC-MS analysis, in vitro anti-oxidant and Vipasha, S., Hardeep, K., Tarun, K., Tullika, M., 2016. Traditional Indian herb
anti-microbial activity of the Catharanthus roseus and Moringa oleifera leaf extracts. Catharanthus roseus used as cancer treatment: a review. Int. J. Pharmacogn.
Data in Brief 29, 105258. Phytochem. Res. 8, 1926–1928.
Tabuti, J.R., Lye, K.A., Dhillion, S.S., 2003. Traditional herbal drugs of Bulamogi, Vutukuri, V.R., Das, M.C., Reddy, M., Prabodh, S., Sunethri, P., 2017. Evaluation of acute
Uganda: plants, use and administration. J. Ethnopharmacol. 88, 19–44. oral toxicity of ethanol leaves extract of Catharanthus roseus in Wistar Albino Rats.
Tam, M.N., Nikolova-Damyanova, B., Pyuskyulev, B., 1995. Quantitative thin layer J. Clin. Diagnostic Res. JCDR 11, 1–4.
chromatography of indole alkaloids. II. Catharanthine and vindoline. J. Liq. Wang, C.H., Wang, G.C., Wang, Y., Zhang, X.Q., Huang, X.J., Ye, W.C., 2012a. Three new
Chromatogr. R.T 18, 849–858. monomeric indole alkaloids from the roots of Catharanthus roseus. J. Asian Nat.
Tang, C., Li, L., Ma, X., Wang, J., Chen, B., Dai, X., Zhang, Y., Chen, X., 2020. Qualitative Prod. Res. 14, 249–255.
and quantitative determination of anaprazole and its major metabolites in human Wang, C.H., Wang, G.C., Wang, Y., Zhang, X.Q., Huang, X.J., Zhang, D.M., Chen, M.F.,
plasma. J. Pharmaceut. Biomed 183, 113146. Ye, W.C., 2012b. Cytotoxic dimeric indole alkaloids from Catharanthus roseus.
Tang, Z., Rao, L., 2010. Applying aluminum oxide column chromatography purify Fitoterapia 83, 765–769.
extracts of cathuranthus roseus and simultaneous determination of the indole Wang, C.H., Zhang, Y., Jiang, M.M., 2014. Indole alkaloids from the roots of Catharanthus
alkaloids by HPLC. Anal. Lett. 43, 219–225. roseus. Chem. Nat. Compd. 49, 1177–1178.
Tang, Z., Yang, L., Zu, Y., Guo, X., 2009. Variations of vinblastine accumulation and Wang, Z., Yang, Z., Zhang, L., Wang, X., Peng, C., Meng, D., Yan, D., 2017. Toxic effects
redox state affected by exogenous H2O2 in Catharanthus roseus (L.) G. Don. Plant of vinca alkaloids from Catharanthus roseus (L.) G. Don on Tetrahymena thermophila
Growth Regul 57, 15–20. BF5 growth by microcalorimetry. Thermochim. Acta 651, 53–57.
Thabet, I., Guirimand, G., Guihur, A., Lanoue, A., Courdavault, V., Papon, N., Bouzid, S., Wesołowska, A., Grzeszczuk, M., Wilas, J., Kulpa, D., 2016. Gas chromatography-mass
Giglioli-Guivarc’h, N., Simkin, A.J., Clastre, M., 2012. Characterization and spectrometry (GC-MS) analysis of indole alkaloids isolated from Catharanthus roseus
subcellular localization of geranylgeranyl diphosphate synthase from Catharanthus (L.) G. don cultivated conventionally and derived from in vitro cultures. Not. Bot.
roseus. Mol. Biol. Rep. 39, 3235–3243. Horti Agrobot. Cluj-Napoca 44, 100–106.
Thakore, D., Srivastava, A.K., Sinha, A.K., 2015. Model based fed batch cultivation and Widowati, W., Mozef, T., Risdian, C., Ratnawati, H., Tjahjani, S., Sandra, F., 2011. The
elicitation for the overproduction of ajmalicine from hairy roots of Catharanthus comparison of antioxidative and proliferation inhibitor properties of Piper betle L.,
roseus. Biochem. Eng. J. 97, 73–80. Catharanthus roseus (L.) G. Don, Dendrophtoe petandra L., Curcuma mangga Val.
Thakore, D., Srivastava, A.K., Sinha, A.K., 2013. Yield enhancement strategies for extracts on T47D cancer cell line. Int. Res. J. Biochem. Bioinform 1, 22–28.
enhancement of indole alkaloids in hairy root cultures of Catharanthus roseus. Int. J. Widowati, W., Mozef, T., Risdian, C., Ratnawati, H., Tjahyani, S., Sandra, F., 2010.
Chem. Eng. Appl. 4, 153–156. Apoptosis and antioxidant activities of Catharanthus rosues (L.) G. Don extract on
Thiäner, J.B., Nett, L., Zhou, S., Preibisch, Y., Hollert, H., Achten, C., 2019. Identification breast cancer cell line. IJCC 1, 99–107.
of 7–9 ring polycyclic aromatic hydrocarbons in coals and petrol coke using High Wiernikowski, J.T., Bernhardt, M.B., 2020. Review of nutritional status, body
performance liquid chromatography–Diode array detection coupled to Atmospheric composition, and effects of antineoplastic drug disposition. Pediatr. Blood Canc.
pressure laser ionization–Mass spectrometry (HPLC-DAD-APLI-MS). Environ. Pollut. 2020, 1–11.
252, 723–732. Wittkamp, B.L., Hawthorne, S.B., Tilotta, D.C., 1997. Determination of aromatic
Thompson, J.E., 2017. Parts-per-billion limits of detection via absorbance spectroscopy: compounds in water by solid phase microextraction and ultraviolet absorption
an ultraviolet (254 nm) absorbance detector for liquid chromatography using a light spectroscopy. 1. Methodology. Anal. Chem. 69, 1197–1203.
emitting diode (LED). Eurasian J. Anal. Chem. 12, 901–911. Wong, P.L., Royce, A.J., Lee-Parsons, C.W., 2004. Improved ajmalicine production and
Tikhomiroff, C., Jolicoeur, M., 2002. Screening of Catharanthus roseus secondary recovery from Catharanthus roseus suspensions with increased product removal rates.
metabolites by high-performance liquid chromatography. J. Chromatogr. A 955, Biochem. Eng. J. 21, 253–258.
87–93. Yamamoto, K., Takahashi, K., Mizuno, H., Anegawa, A., Ishizaki, K., Fukaki, H.,
Ohnishi, M., Yamazaki, M., Masujima, T., Mimura, T., 2016. Cell-specific

43
S. Kumar et al. Journal of Ethnopharmacology 284 (2022) 114647

localization of alkaloids in Catharanthus roseus stem tissue measured with Imaging Zhou, H., Tai, Y., Sun, C., Pan, Y., 2005. Rapid identification of vinca alkaloids by direct-
MS and Single-cell MS. Proc. Natl. Acad. Sci. Unit. States Am. 113, 3891–3896. injection electrospray ionisation tandem mass spectrometry and confirmation by
Yang, J., Guo, J., Yuan, J., 2008. In vitro antioxidant properties of rutin. LWT - Food Sci. high-performance liquid chromatography–mass spectrometry. Phytochem. Anal. 16,
Technol. (Lebensmittel-Wissenschaft -Technol.) 41, 1060–1066. 328–333.
Yang, L., Wang, H., Zu, Y.G., Zhao, C., Zhang, L., Chen, X., Zhang, Z., 2011. Ultrasound- Zhou, P., Yang, J., Zhu, J., He, S., Zhang, W., Yu, R., Zi, J., Song, L., Huang, X., 2015.
assisted extraction of the three terpenoid indole alkaloids vindoline, catharanthine Effects of β-cyclodextrin and methyl jasmonate on the production of vindoline,
and vinblastine from Catharanthus roseus using ionic liquid aqueous solutions. Chem. catharanthine, and ajmalicine in Catharanthus roseus cambial meristematic cell
Eng. 172, 705–712. cultures. Appl. Microbiol. Biotechnol. 99, 7035–7045.
Yu, B., Pan, Y., Liu, Y., Chen, Q., Guo, X., Tang, Z., 2021. A comprehensive analysis of Zhu, J., He, S., Zhou, P., Zi, J., Liang, J., Song, L., Yu, R., 2018. Eliciting effect of
transcriptome and phenolic compound profiles suggests the role of flavonoids in catharanthine on the biosynthesis of vallesiachotamine and isovallesiachotamine in
cotyledon greening in Catharanthus roseus seedling. Plant Physiol. Biochemist 167, Catharanthus roseus cambial meristematic cells. Nat. Prod. Commun 13,
185–197. 1934578–1801300508.
Zárate, R., Verpoorte, R., 2007. Strategies for the genetic modification of the medicinal Zhu, J., Wang, M., Wen, W., Yu, R., 2015. Biosynthesis and regulation of terpenoid indole
plant Catharanthus roseus (L.) G. Don. Phytochem. Rev. 6, 475–491. alkaloids in Catharanthus roseus. Phcog. Rev. 9, 24–28.
Zhang, L., Qing-Hui, G.A.I., Yuan-Gang, Z.U., Lei, Y.A.N.G., Yu-Liang, M.A., Yang, L.I.U.,
2014. Simultaneous quantitative determination of five alkaloids in Catharanthus
roseus by HPLC-ESI-MS/MS. Chin. J. Nat. Med. 12, 786–793. Further reading
Zhang, L., Wei, G., Liu, Y., Zu, Y., Gai, Q., Yang, L., 2016. Antihyperglycemic and
antioxidant activities of total alkaloids from Catharanthus roseus in streptozotocin- Integrated Taxonomic information system- ITIS https://www.itis.gov/servlet/SingleRpt/
induced diabetic rats. J. For. Res. 27, 167–174. SingleRpt?search_topic=TSN&search_value=30168#nullsearch_topic=TS
Zhang, W., Yang, J., Zi, J., Zhu, J., Song, L., Yu, R., 2015. Effects of adding vindoline and Nandsearch_value=30199#null (2020) (accessed on 12th September 2020).
MeJA on production of vincristine and vinblastine, and transcription of their POWO, 2021. Plants of the World Online. Facilitated by the Royal Botanic Gardens, Kew.
biosynthetic genes in the cultured CMCs of Catharanthus roseus Nat. Prod. Commun. Lee-Parsons, C.W., Royce, A.J., 2006. Precursor limitations in methyl jasmonate-induced
Now. 10, 1934578–1501001220. Catharanthus roseus cell cultures. Plant Cell Rep. 25, 607–612.
Zhang, W.K., Xu, J.K., Tian, H.Y., Wang, L., Zhang, X.Q., Xiao, X.Z., Li, P., Ye, W.C., Liu, D.H., Ren, W.W., Cui, L.J., Zhang, L.D., Sun, X.F., Tang, K.X., 2011. Enhanced
2013. Two new vinblastine-type N-oxide alkaloids from Catharanthus roseus. Nat. accumulation of catharanthine and vindoline in Catharanthus roseus hairy roots by
Prod. Res. 27, 1911–1916. overexpression of transcriptional factor ORCA2. Afr. J. Biotechnol. 10, 3260–3268.
Zhang, X.N., Liu, J., Liu, Y., Wang, Y., Abozeid, A., Yu, Z.G., Tang, Z.H., 2018. Magnotta, M., Murata, J., Chen, J., De Luca, V., 2006. Identification of a low vindoline
Metabolomics analysis reveals that ethylene and methyl jasmonate regulate different accumulating cultivar of Catharanthus roseus (L.) G. Don by alkaloid and enzymatic
branch pathways to promote the accumulation of terpenoid indole alkaloids in profiling. Phytochemistry 67, 1758–1764.
Catharanthus roseus. J. Nat. Prod. 81, 335–342. O’Gara, J.E., Walsh, D.P., Alden, B.A., Casellini, P., Walter, T.H., 1999. Systematic study
Zhang, Y., Sun, Y., Mu, X., Yuan, L., Wang, Q., Zhang, L., 2017. Identification of of chromatographic behavior vs alkyl chain length for HPLC bonded phases
metabolites of vindoline in rats using ultra-high performance liquid containing an embedded carbamate group. Anal. Chem. 71, 2992–2997.
chromatography/quadrupole time-of-flight mass spectrometry. J. Chromatogr. B Qu, Y., Safonova, O., De Luca, V., 2019. Completion of the canonical pathway for
1060, 126–137. assembly of anticancer drugs vincristine/vinblastine in Catharanthus roseus. Plant J.
Zhao, J., Verpoorte, R., 2007. Manipulating indole alkaloid production by Catharanthus 97, 257–266.
roseus cell cultures in bioreactors: from biochemical processing to metabolic Shanks, J.V., Bhadra, R., Morgan, J., Rijhwani, S., Vani, S., 1998. Quantification of
engineering. Phytochemistry Rev. 6, 435–457. metabolites in the indole alkaloid pathways of Catharanthus roseus: implications for
Zhao, J., Zhu, W.H., Hu, Q., 2001. Enhanced catharanthine production in Catharanthus metabolic engineering. Biotechnol. Bioeng. 58, 333–338.
roseus cell cultures by combined elicitor treatment in shake flasks and bioreactors. Singh, A.K., Singh, S., Srivastava, M., Srivastava, S., 2008. Seed and seedling morphology
Enzyme Microb. Tech 28, 673–681. of a medicinal plant-Catharanthus roseus. Indian For. 134, 537–541.
Zhao, L., Sander, G.W., Shanks, J.V., 2013. Perspectives of the metabolic engineering of St-Pierre, B., De Luca, V., 1995. A cytochrome P-450 monooxygenase catalyzes the first
terpenoid indole alkaloids in Catharanthus roseus hairy roots. Biotechnology of Hairy step in the conversion of tabersonine to vindoline in Catharanthus roseus. Plant
Root Systems (pp. 23–54). Springer, Berlin, Heidelberg. Physiol. 109, 131–139.
Zhao, Y., Nookandeh, A., Schneider, B., Sun, X., Schmitt, B., Stöckigt, J., 1999. Lignans The Plant List, 2013. Version 1.1. Published on the Internet (accessed 1st January). htt
from Torreya jackii identified by stopped-flow high-performance liquid p://www.theplantlist.org/.
chromatography–nuclear magnetic resonance spectroscopy. J. Chromatogr. A 837, Verpoorte, R., Niessen, W.M.A., 1994. Liquid chromatography coupled with mass
83–91. spectrometry in the analysis of alkaloids. Phytochem. Anal. 5, 217–232.
Zheng, Z., Wu, M., 2004. Cadmium treatment enhances the production of alkaloid Zheng, W., Wang, S.Y., 2001. Antioxidant activity and phenolic compounds in selected
secondary metabolites in Catharanthus roseus. Plant Sci. 166, 507–514. herbs. J. Agric. Food Chem. 49, 5165–5170.

44

You might also like