Top Banner
1 Review Article Medicinal Plants Sources of Anticancer Drugs Latifa Doudach 1,2 , Bouchra Meddah 1 , Laila Benbacer 3 , Layachi Chabraoui 4 , My.A. Faouzi 1 , Abdelhakim Elomri 2 and, Yahia Cherrah 1 1 Mohammed V Souissi University of Rabat, Faculty of Medicine and Pharmacy, Laboratory of Pharmacology and Toxicology, Research Team pharmacokinetic, Morocco 2 University of Rouen, CNRS UMR 6014, C.O.B.R.A, UFR Medicine and Pharmacy, 22 Boulevard Gambetta 76183 Rouen, France 3 Biology Unit and Medical Research CNESTEN, PB 1382 RP, 10001 Rabat, Morocco 4 Biochemical laboratory, Ibn Sina Hospital Rabat, Morocco. *Corresponding authors: Pr. Bouchra Meddah, laboratory of Pharmacology and Toxicology, Faculty of Medicine and Pharmacy, Mohammed V Souissi University, Rabat, Morocco. Tel.: +212 - 5 37 77 04 21, Fax: +212 5 37 77 37 01 E-mail address: [email protected]
39
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
  • 1

    Review Article

    Medicinal Plants Sources of Anticancer Drugs

    Latifa Doudach1,2

    , Bouchra Meddah1, Laila Benbacer

    3, Layachi Chabraoui

    4, My.A.

    Faouzi1, Abdelhakim Elomri

    2 and, Yahia Cherrah

    1

    1Mohammed V Souissi University of Rabat, Faculty of Medicine and Pharmacy, Laboratory of

    Pharmacology and Toxicology, Research Team pharmacokinetic, Morocco

    2University of Rouen, CNRS UMR 6014, C.O.B.R.A, UFR Medicine and Pharmacy, 22

    Boulevard Gambetta 76183 Rouen, France

    3Biology Unit and Medical Research CNESTEN, PB 1382 RP, 10001 Rabat, Morocco

    4Biochemical laboratory, Ibn Sina Hospital Rabat, Morocco.

    *Corresponding authors: Pr. Bouchra Meddah, laboratory of Pharmacology and Toxicology,

    Faculty of Medicine and Pharmacy, Mohammed V Souissi University, Rabat, Morocco. Tel.:

    +212 - 5 37 77 04 21, Fax: +212 5 37 77 37 01

    E-mail address: [email protected]

  • 2

    Abstract

    Medicinal plants continue to generate interest in pharmacological research and development of

    new anticancer agent. Approximately 64% of drugs used in cancer chemotherapy are from

    plants materials with different modes of action. This study proposes to list the natural

    compounds having the capacity to inhibit or reduce the development of cancer in order to

    identify their mechanisms of action and mode of signalization involved in the process of

    carcinogenesis, vinblastine, vincristine, topotecan, irinotecan, etoposide, taxol and paclitaxel

    are drugs derived from natural resources used in the treatment of a wide variety of cancers.

    Data on safety and efficacy are available for an even few number of plants.

    Keywords: Medicinal plants, natural products, drugs, cancer.

  • 3

    1. Introduction

    Remedies made from plants constitute a promising avenue for the development of drugs

    traditionally improved. Estimations from the World Health Organization (WHO) say that over

    80% of the population use traditional medicine in health care needs. In fact about 35000 to

    70000 plant species are used for medical purposes in the world [1]

    and over 62% of new

    biologically active substances are currently used as anticancer agents made from natural

    sources (plants, marine organisms and micro-organizations) [2]

    . Studies of separation and

    chemical identification have revealed the pharmacological potential of several medicinal plants

    [3,4,5]. The plants are then a source of natural substances with great potential of application and

    whose physicochemical properties seem to be related to their compositions of secondary

    metabolites. Different therapeutic effects have been associated with natural compounds and

    have allowed the isolation of several phytoactive compounds (polyphenols, terpenods,

    alkaloids, glycosides and polysaccharides ...). Several studies have documented the use of

    plants in the Ethnotherapy and the treatment of several diseases. The root bark extracts of

    Calotropis procera, have been the target in multiple uses in Burkina Faso, have demonstrated

    antitumor activity [6]

    . Several studies have reported the use of plants in the treatment of

    Ethnotherapy and other diseases. Hartwell has estimated the use of over 3 000 plant species in

    the treatment of cancer [7]

    . Several chemotherapy drugs are isolated from medicinal plants,

    including the Vinca alkaloids, vinblastine and vincristine, isolated from the plant of

    Catharanthus roseus, etoposide and teniposide, semisyntheticaly derivative from

    epipodophyllotoxin isomer of podophyllotoxin (isolated from kind of Podophyllum species),

    taxanes isolated from Taxus species, the semisynthetic derivatives of camptothecin: topotecan

    and irinotecan, isolated from the plant Camptotheca acuminata [8,9,10]

    . Given the current

    worldwide interest aroused by the use of medicinal plants to fight disease and maintain health,

    knowledge of these plants is now essential and is even crucial in health sector and

  • 4

    pharmaceutical industry. Nature is a source not only of potential chemotherapeutic agents and

    lead compounds that have provided the basis and inspiration for the semisynthesis or total

    synthesis of effective new drugs, half of the existing pharmaceuticals today are inspired by

    natural products [11,12]

    . The improvement of medicinal plants value can be achieved by

    searching newer, more effective and less toxic therapeutic molecules, which will add great

    values to the resources that can be later, integrated into the therapeutic arsenal already in use.

    This work represents a literature review exposing the interest of the phytoactive isolated from

    medicinal plants used as active principles for chemotherapy. The identified active extracts are

    standardized, will bear the different preclinical steps, pharmaceutical, analytical, toxicological,

    pharmacological and clinical according to current regulations. The finished products will be

    subject to authorization on the market (AMM) for its efficacy, safety and pharmaceutical

    quality. Therefore, the effectiveness of anticancer activity phytoactifs to is reproducible to the

    modern medicine, the doses are standardized contrary to traditional medicine, the active

    ingredients from plants in complex mixtures, undefined, cant generate a comparable activity.

    2. Medicinal Products Made from Aromatic and Medicinal Plants

    Most current cancer therapies involve the modulation of a single target. The high cost of

    conventional drugs and the desire to consume unprocessed agricultural products, encouraged

    the development of several alternative approaches. The use of natural remedies, made from

    plants, remains a very promising way to address health problems, such as cancer and other

    associated pathologies. Actually, several drugs derived from natural sources are used in

    chemotherapy (Table I), among the large classes of anticancer drugs made from plants [13]

    we

    find:

    2.1. Podophyllotoxin: Podophyllotoxin (1), isolated from plants of the Berberidaceae family:

    Podophyllum peltatum and Podophyllum hexandrum has given birth after a few structural

    changes, to three compounds: Teniposid (2), Etoposid (3) and phosphate of etoposid widely

  • 5

    used in antitumor therapy [14]

    , it is used against several types of tumors, including lung cancer,

    testicular, lymphoma, leukemia, sarcoma and Kaposi... the mechanism of action of

    podophyllotoxin is based on the inhibition of tubulin polymerization, which causes the

    inhibition of tubulin polymerization and hence microtubule assembly, thus arresting the cell

    cycle in metaphase [15,16]

    . The podophyllotoxin derivatives induce apoptosis in proliferating

    cells by interacting with the topoisomerase II, homodimeric enzyme controlling the degree of

    supercoiling of deoxyribonucleic acid (DNA), the etoposid increases the number of double-

    stranded cuts on DNA in proliferating cells [17]

    . Alterations in DNA structure are associated

    with activation of a protein kinase, ATM (ataxia telangiectasia mutated kinase, which

    stimulates the active form of the tumor suppressor protein p53, leading to the cell cycle arrest

    (en G2) and / or to apoptosis [18]

    .

  • 6

    Podophyllotoxin (1) Podophyllotoxin (1)

    Teniposid (2)Teniposid (2)

    Etoposid (3)Etoposid (3)

    2.2. Vinca alkaloids: The isolation of Vinca: vinblastine (4) and vincristine (5) from the

    Madagascar periwinkle, Catharanthus roseus G. Don. (Apocynaceae) has introduced a new era

    of using plant material as anticancer agents, these phytoactive are used in combination for the

    treatment of cancer, including cancer of leukemia, lymphomas, testicular, breast, lung, and

    Kaposi's sarcoma [19]

    . The semi-synthetic analogues are vinorelbine (6) and vindesine (7),

    obtained from vindoline and catharanthine, which play beneficial role for the treatment of

    breast and lung cancers. Vinblastine and vincristine are the first antimitotic identified drugs

    causing a malformation of the mitotic spindle [20]

    . They targeted the subunit of tubulin and

    depolymerized microtubules.

  • 7

    Vinblastine (4)Vinblastine (4)

    Vincristine (5)Vincristine (5)

    Vinorelbine (6)Vinorelbine (6)

    Vindesine(7)Vindesine(7)

  • 8

    Vinblastine and vincristine are commonly used in the treatment of some cancers (testicular

    cancer, Hodgkin's disease, acute lymphocytic leukemia). Vinblastine binds to tubulin, the

    protein of microtubules of the spindle. It inhibits the assembly of microtubules, resulting in

    dissolution of the mitotic spindle and the cell is then is arrested in metaphase. In fact, by

    binding to tubulin, vinblastine prevents some of his contacts to settle; thus, the lateral contacts

    between protofilaments, which are crucial for the stability of microtubules, cannot be

    established. Microtubules are hollow cylinders whose walls are made of polymers (or

    profilements) of tubulin, an abundant cellular heterodimeric protein consisting of an subunit

    and a subunit. A high concentration of vinblastine, when the loss of microtubular contacts

    becomes important, the tubulin assembles in spirals at the expense of the formation of

    microtubules. In these spirals, the link between tubulin heterodimers is enhanced by contact

    with vinblastine molecules, each molecule of vinblastine interacts with the subunit of a

    heterodimer and the subunit of the other forming a antimitotic complex [21].

    2.3. Camptothecin: Isolated in the early 1970's from the extract of the bark of the tree

    Camptotheca acuminata (Nyssaceae), an ornamental tree from China, the camptothecin (8) is

    highly toxic for clinical usage. It belongs to the category of anticancer drugs that inhibit

    topoisomerase I, a nuclear enzyme that allows supercoiled DNA to relax, thus, allowing

    replication, recombination, transcription and DNA repair [22]

    . The effect of camptothecin is to

    stabilize a cleavable complex (ADNtopI) formed at many sites on the double helix. Once

    stabilized, the complex stops the replication fork, which, in turn, leads to inhibition of DNA

    synthesis and promotion of cell death. Camptothecin is a potent inhibitor of the transcription of

    ribosomal and messenger RNA. This inhibition is mainly due to blockage of the elongation by

    trapping the cleavage complex Topi / DNA. In studies conducted on dhydrofolate reductase

    (DHFR) in Chinese hamster, it appears that camptothecin activates the initiation, but inhibits

    the elongation of gene transcription. In human cells, inhibition of transcription by camptothecin

  • 9

    is not uniform. While it strongly inhibits the promoter of the endogenous gene c-MYC,

    camptothecin has a minimal effect on the episomal gene c-MYC or on the basal transcription of

    HSP70 genes and glyceraldehyde 3-phosphate dehydrogenase (GAPDH). The inhibition of the

    catalytic activity of the Topoisomerase can also prevent transcription by appearance and

    accumulation of supercoiled upstream of the transcription complex RNA polymerase.

    Topotecan (9) and irinotecan (10) are the synthetic analogues of camptothecin. Topotecan is

    used clinically for the treatment of ovarian and small cell lung cancers. Once topoisomerase I

    (top I) covalently linked to double-stranded DNA, the binding of topotecan mimics a DNA

    base by being inserted at the cleavage site [23]

    . This interaction changes the position of the free

    end of the 5'-OH DNA strand cut by banning a subsequent religation. Irinotecan in turn is used

    clinically for the treatment of colorectal cancer; the stability of the lactone nucleus of irinotecan

    is about 20 times higher than that of camptothecin [24]

    , its action is to prevent the reconstitution

    of stranded DNA after cleavage, thereby inhibiting the correct synthesis of DNA. Indeed,

    irinotecan binds to the cleavage complex formed by the top I-DNA and inhibits the religation

    of DNA fragments.

  • 10

    Camptothecin (8) Camptothecin (8)

    N(CH3)2

    Topotecan (9)

    N(CH3)2

    Topotecan (9)

    Irinotecan (10)Irinotecan (10)

    2.4. Taxoids: The structure of paclitaxel (11) has been elucidated in 1971 and was introduced in

    the clinic (U.S market) in the 1990s [25,26]

    this molecule was isolated from the yew (Taxus

    baccata). The Paclitaxel acts by inducing microtubule polymerization and inhibiting their

    depolymerization (subunit alpha and beta). This mechanism of action leads to a mitotic arrest in

    metaphase-anaphase stage, leading to the inhibition of proliferation and promotion of cell death

    [27]. At low concentrations, cell death occurs after an aberrant mitosis, involving the p53

    protein. Once p53 is activated, it acts as a transcription factor that regulates the expression of

    many components involved in the pathways of cell cycle regulation. The p21WAF1/CIP1

    protein, mostly known for its role as an inhibitor of CDK and kinas proteins, plays also a key

    role in the regulation of the cell cycle progression [28]

    . P53 the gatekeeper of the genome,

    induces the transcription of the p21WAF1/CIP1 protein that blocks the passage of the cell from

    G1 to S phase by inhibiting cyclin D-cdk complex. At high concentrations, there is a mitotic

    arrest in G2 / M phase. The accumulation of the mass of microtubules in cells and the induction

  • 11

    of apoptosis through mitogen-activated protein kinase (MAP kinase) or protein kinase A are

    able to induce bcl2 phosphorylation by inhibiting its anti-apoptotic action [29.30]

    . Paclitaxel is

    significantly active against ovarian cancer, advanced breast cancer and lung cancer [26]

    . The

    semi-synthetic analogue of paclitaxel is docetaxel (12).

    Paclitaxel (11)Paclitaxel (11)

    Docetaxel (12)Docetaxel (12)

    2.5. Colchicine: Colchicine (13), a tricyclic alkaloid isolated in 1820 by PJ Pelletier and JB

    Caventou from plants of Gloriosa superba L and Colchicum autumnale, induces cell death by

    arresting mitosis in metaphase by inhibiting tubulin polymerization, the formation and function

    of microtubules, and by forming combinations with tubulin [31]

    . Three proteins play a key role

    in the pharmacokinetics of colchicine: tubulin, its elective target; the cytochrome CYP3A4,

    which is involved in its metabolism and ABCB1 protein, which regulates its tissue distribution,

    and renal and biliary excretion [32]

    . Colchicine has a direct effect on P-glycoprotein, whose

    particularity is to destroy tumor vasculature, causing their necrosis [33]

    . Effective doses of

    phytoactive are close to the maximum tolerated dose; it induces strong toxicity and significant

    morbidity. Colchicine analogues are obtained by substituting one or more radicals, giving

  • 12

    comparable efficacy and less toxicity: the N-desacetyl-colchicine (14), the N-desacetyl-N-

    methyl-colchicine (15), trimethyl-colchicine acid and colchicoside (16).

    R1 = COCH3; R2 = Me Colchicine (13)R1 = H; R2 = Me N-desacetyl-colchicine (14)R1 = CH3; R2 = Me N-desacetyl- N-methyl colchicine (15)R1 = COCH3; R2 = Glu Colchicoside (16)

    R1 = COCH3; R2 = Me Colchicine (13)R1 = H; R2 = Me N-desacetyl-colchicine (14)R1 = CH3; R2 = Me N-desacetyl- N-methyl colchicine (15)R1 = COCH3; R2 = Glu Colchicoside (16)

    3. Phytoactifs with Anticancer Activity

    Natural substances allow the synthesis of selective analogues with increased biological

    properties [34,35]

    , many phytoactive isolated from plants are currently in clinical and preclinical

    trials.

    3.1. Betulinic acid: The betulinic acid (17), which is a pentacyclic triterpene obtained from

    many vegetal or synthetic species from betulin, a substance found abundantly in certain species

    of birch (Betula papyrifera). It displays anti-tumor activity vis--vis the tumor cell lines and

    against other melanocytic tumors ectodermal Such as neuroblastoma, glioma, medulloblastoma

    and Ewing sarcoma [36]

    . The mechanism of action of betulinic acid is mainly manifested by the

    induction of apoptosis through proteolytic cleavage of caspases 3 and 8, which is preceded by

    the appearance of mitochondrial events and the generation of reactive substances of oxygen,

    involving an apoptotic pathway and a mitochondrial phase [37]

    . The betulinic acid has

    interesting therapeutic and anticancer activities. It inhibits the enzyme involved in the

    mechanism of DNA repair. Thus, the activity of bleomycin (an antitumor agent) that acts by

    damaging the DNA of cancer cells is accentuated in the presence of betulinic acid [38]

    .

  • 13

    Betulinic acid (17) Betulinic acid (17)

    3.2. Curcumin: Curcumin or diferuloylmethane (18) isolated from the plant Curcuma longa L.

    plays interesting roles in the treatment of cancer; moreover, it has an anti-angiogenic action,

    which could explain its chemopreventive effect [39]

    . The anticancer potential of curcumin

    results from its ability to stop the proliferation and metastasis of a variety of tumor cells by

    inhibiting adhesion molecules and consequently angiogenesis, a cellular process and a crucial

    step in the growth and the metastasis of many cancers. This polyphenol inhibit also the activity

    of cyclin D1, a proto-oncogene overexpressed in many cancers at higher concentrations.

    Curcumin induces apoptosis by blocking the effect of proteins that regulate this process and by

    modulating transcription factors. Human clinical trials indicated no dose-limiting toxicity (up to

    10 g / day) and a huge anti-cancer potential of curcumin demonstrated in vitro (in italics) on

    various tumor cell lines [40]

    .

    Curcumin (18) Curcumin (18)

    3.3. Lapachol: The Lapachol (19) is a phytoactive isolated from avellanedae Tabebuia

    (Bignoniaceae), tree of South Africa. Highly active against a variety of cancer cells, it blocks

    tumor growth at micromolar doses and inhibits invasion and metastasis of cancer cells by

  • 14

    altering the protein profiles of invasive cells. The cytotoxicity of this active compound on

    hepatocellular carcinoma (HepG2) is significant, and induces apoptosis of these cells by

    caspase activation [41.42]

    . Topoisomerase I and II are the elective targets of this phytoactive, that

    effective for the treatment of many types of cancer (breast, prostate and pancreas).

    Lapachol (19) Lapachol (19)

    3.4. Ellipticine: Ellipticine (5,11-dimethyl-6H-pyrido (4,3-b) carbazole) (20), an alkaloid

    isolated from plants of Ochrosia borbonica, Excavatia coccinea and Ochrosia elliptica from

    the family of Apocynaceae, used in cancer chemotherapy for the treatment of a wide variety of

    cancers. The main mechanism of action of ellipticine is the inhibition of topoisomerase II.

    Ellipticine has the ability to bind to proteins and induce the cytotoxic harmful free radicals to

    the body [43]

    ; moreover, Ellipticine inhibit the cytochrome P4501A1 [44]

    and activate the

    transcription of the mutant protein P53. Recently, it has been shown that ellipticine derivatives

    can induce stress of the endoplasmic reticulum of cells, and suggest that stress is a contributing

    factor to the cytotoxic activity of this class of inhibitors of topoisomerase II. This phytoactive

    inhibits cell growth and induces apoptosis of cancer cells of hepatocellular carcinoma (HepG2)

    [45].

    Ellipticine (20) Ellipticine (20)

  • 15

    3.5. Combretastatin A4: Combretastatin A4 (CA4) (21), isolated from the bark of a South

    African tree Combretum caffrum, is an antimitotic agent in cancer cells and selectively inhibits

    tubulin polymerization of tumor endothelial cells [46]

    . Indeed, disruption of microtubule

    cytoskeleton in the CA4 inhibits cell division of endothelial cells in the proliferative phase and

    induces apoptosis of this latter. This product induces cell cycle arrest in the growth phase (G2)

    or mitosis (M) and acts on the cell proliferation phase and not on the cell quiescent phase [47]

    .

    Combretastatin A4 also acts on the vasculature of solid tumors. It induces the specific

    destruction of blood vessels and inhibit the formation of new vessels or anti-angiogenesis [48]

    .

    The phenstatin (22) is an analogue of the CA4 and whose cytotoxic activity is very important.

    The CA4 is currently in Phase III of clinical trials as a phosphate product. The combretastatin

    A4 phosphate (CA4-P) is an antitubulin agent used for its anti-angiogenic and antivascular

    properties.

    Combretastatin A4 (21) Combretastatin A4 (21)

    Phenstatin (22) Phenstatin (22)

    3.6. Flavopiridol: Flavopiridol (23), a semi-synthetic compound derived from the rohitukine

    (24), is an alkaloid isolated from the bark of a plant widely used in India, Dysoxylum

    binectariferum. Flavopiridol acts by inhibiting the cyclin-dependent kinase (cdk 1, 2 and 4) [49]

    .

    Indeed, it binds competitively to the receptors of adenosine triphosphate (ATP), necessary for

    the activation of cyclin-dependent kinases (CDKs) and inhibits the phosphorylation of the

    amino acid threonine of Cdks (this phosphorylation is required for the activation of Cdks).

    Flavopiridol induces apoptosis of several cell lines; furthermore, it reduces the levels of cyclin

  • 16

    D1 and inhibit angiogenesis [50,51]

    through a decrease in the matrix metalloproteinases secretion

    leading to the inhibition of c-erbB-2 gene expression. Overexpression of this gene (c-erbB-2) in

    breast cancers is correlated with invasion and metastatic progression of these tumors [52]

    through alteration of the stability of ribonucleic acids (mRNAs) encoding the vascular

    endothelial growth factor (VEGF), which represent a pathophysiological stimulus for the

    induction of angiogenesis in vivo [53]

    . Flavopiridol acts synergistically with several

    conventional antitumor drugs. It potentiates the proapoptotic effect of Mitomycin C and

    paclitaxel by having a chemopreventive effect of chromosomal abnormalities and genetic

    instability induced by paclitaxel [54]

    . Flavopiridol is currently in Phase I and II of clinical trials,

    and it is used for the treatment of a wide variety of cancers, including breast, stomach,

    leukemia, lymphomas and solid tumors [55]

    .

    Flavopiridol (23) Flavopiridol (23)

    Rohitukine (24) Rohitukine (24)

    3.7. Roscovitine: Roscovitine (25), a synthetic derivative of olomoucine (26), is a natural

    product isolated from the plant Raphanus sativus L. Roscovitine initiates the blockage of the

    cell cycle in phase G0, G1 and S and in the transition phase G2 / M, leading to the inhibition of

    CDK / cyclin complexes, a protein involved in the regulation of cell cycle [56]

    . Roscovitine

    inhibits the activity of CDK7, which in turn inhibits the phosphorylation of threonine of CDK1

    2 and 4, leading to the prevention of the catalytic activity of the CDK2/cyclineE complex. This

    inactivation will lead to a lack of phosphorylation required for the degradation of the natural

    inhibitor p27KIP1, whose stability helps in maintaining its role as an inhibitor of CDK2 and

  • 17

    CDK4 and thus blocking the cell cycle in G1. Roscovitine is also responsible for stopping cell

    division through the deregulation of the activity of CDKs, leading to the blockage of DNA

    replication, the formation of the nucleolus, the duplication of centrosome, the fragmentation of

    the Golgi apparatus and finally the rupture of the nuclear envelope, thus, leading to cell death,

    characterized by caspase activation and cytochrome C release. This product is used to treat

    breast and lung cancers. In Europe, roscovitine is in phase II in clinical trials [19, 57]

    .

    Roscovitine (25) Roscovitine (25)

    Olomoucine (26) Olomoucine (26)

    3.8. Daphnoretin: Daphnoretin (27) belongs to the class of bicoumarines, mainly found in

    Thymelaeaceae, but also in legumes and Rutaceae [58]

    . Historically, daphnoretin (3.7 '-

    dicoumarylester), is the first of bicoumarine Thymelaeaceae, isolated in 1963 from the berries

    of Daphne mezereum and leaves of Daphnopsis racemosa [59]

    . Subsequently, the same group of

    researchers isolated the 6-glycoside of daphnoretin called daphnorine of Daphne mezereum [60]

    .

    In 1986, Chakrabarti and al. have isolated the Acetyl daphnoretin [61]

    , while qu'ulubelen et al [62]

    identified the dimethyl daphnoretine extracted from Daphne gnidioides Szovits ex Meisson.

    Several biological activities have been attributed to the Daphnoretin, including antiviral

    activity, initiated by the inhibition of the expression of viral antigens (HBsAg) expressed on the

    surface of target cells (human hepatoma Hep B Cells), via a mechanism of activation of the

    protein kinase C (PKC) pathway [63]

    . It was also shown that Daphnorrtin is also able to

    significantly inhibit the growth of cancerous ascites via inhibition of DNA synthesis and

    protein of cancer cells [64]

    . An anticancer activity was also attributed to Daphnoretine. Indeed,

  • 18

    in several tumor models, the daphnortine appears to inhibit the kinase activity of hEGF

    receptors (Human Epidermal Growth Factor receptor). Considering his medical interest, several

    studies are now optimizing the extraction methods of Daphnoretin from different natural

    resources. Wikstroemia indica (L.) is a model of choice that contains a lot that could serve as a

    prototype with important medical and nutritional value [65]

    . These authors confirmed the anti-

    cancer effect of Daphnoretin on cancer cell lines of cervical HeLa, the human lung

    adenocarcinoma cell A549, cells of neuroendocrine carcinoma (NEC) and tumor cells HE-p2

    [65].

    Daphnoretin (27) Daphnoretin (27)

    3.9. Salvicine: The Salvicine (28) is derived from the structurally modified diterpenequinone,

    isolated from Salvia prionitis Hance. The salvicine has been widely described for its antitumor

    activity in vitro and in vivo [66.67]

    . Using several tumor models, the salvicine showed a broad

    spectrum anti MDR (Multi Drug Resistance) and antimetastatic [68.69]

    . Functionally, the

    salvicine inhibits the catalytic activity of a human enzyme, topoisomerase II (htopo II) which

    causes DNA cleavage. The htopo II is highly expressed in proliferating cells and plays a crucial

    role in replication, transcription and organization of chromosomes [70,71,72]

    . Its depletion leads to

    cell death [71.72]

    . Thus, this nuclear enzyme, highly conserved, is an important target for cancer

    chemotherapy. Approximately 50% of drugs used in cancer chemotherapy are the htopo II

    [73,74,75]. The salvicine inhibits htopo II by binding to the ATPase domain of the enzyme,

    competing with ATP and ADP.Competitive inhibition experiments show that blocking ATP in

    a dose-dependent way and competitive to the setting the salvicine the domain of the enzyme

  • 19

    ATP-ase, indicating that the salvicine and ATP use the same binding site on 'htopo II. In tumor

    cells, the salvicine induced gene-specific DNA damage and triggers cell apoptosis by

    modulating the expression of various genes, including-myc, c-fos, c-JNK, c-jun, and mdr1

    [76,77,78].

    Salvicine (28) Salvicine (28)

    4. Conclusion

    The rapid progress made in cell and molecular biology over the last thirty years played a major

    role in better understanding the functioning of cancer cells. As a result, more specific

    treatments have been developed, and it has become possible to target more precisely the

    processes involved in the development, proliferation and survival of tumor cells. A better

    understanding of the characteristics of tumor cells has recently led to the development of more

    targeted treatments, and therefore generally less toxic. This may include conventional cytotoxic

    molecules targeting non-specific molecules expressed on the surface of cancer cells and at less

    degree in normal proliferating cells (DNA, enzymes, microtubules...), or of molecules directed

    against targets specific to cancer cells (oncogenes). At the present time and given the current

    worldwide interest aroused by the use of natural resources, especially medicinal plants used to

    fight disease and maintain health, a knowledge of these plants is now essential and has a major

    importance in health sector and pharmaceutical industry. In medicine, particularly in the field

    of cancer, the use of herbs is increasingly enhanced especially with the excessive use of

    synthetic drugs and awareness of their toxicity, which contributed in oncology, leading to a

  • 20

    favorable reconsideration of the medicinal practices made from natural herbal. Several research

    studies have led to the discovery and development of new active ingredients from natural

    molecules (or derivatives) and several of these compounds are used today in clinical practice.

    However, apart from the cytotoxic effect of new anticancer agents, only the pre-clinical and

    clinical tests that contrasts in their use as new anticancer drugs and thus their integration into

    the existing therapeutic armamentarium. In conclusion, the use of naturally occurring

    molecules in the treatment of cancer has greatly contributed to the improvement of the

    therapeutic efficacy of drugs used today in cancer chemotherapy. Similarly, the

    pharmacognosy, which contributed to the improvement of the knowledge of natural medicines,

    and to the development of this sector, which offers to the modern medicine great potentials for

    progress, based on the discovery and development of new anticancer molecules based on

    natural substances.

    Conflict of Interest Statement

    There is no conflict of interest associated with the authors of this paper, and the fund sponsors

    did not cause any inappropriate influence on this work.

    References

    1. S. Bhattarai, R.P. Chaudhary, C.L. Quave and R. S. L. Taylor. The use of medicinal plants

    in the Trans-Himalayan arid zone of Mustang district, Nepal, Journal of Ethnobiology and

    Ethnomedicine, vol. 6, no.14, 2010.

    2. G.F. Gonzales and L.G. Valerio, Medicinal plants from Peru: a review of plants as potential

    agents against cancer. Anti-Cancer Agents in Medicinal Chemistry, vol. 6, no. 5, pp. 429-444,

    2006.

    3. E. S. Fiala, B. S. Reddy and J. H. Weisburger, Naturally occurring anticarcinogenic

    substances in foodstuffs. Annual Review of Nutrition, vol. 5, pp. 295-321, 1985.

  • 21

    4. L. C. Tapsell, I. Hemphill, L. Cobiac, C. S. Patch, D. R. Sullivan, M. Fenech, S. Enrys, J. B.

    Keogh, P. M. Clifton, P. G. Williams, V. A. Fazio and K. E. Inge, Health benefits of herbs and

    spices: The past, the present, the future. The Medica Journal of Australia, vol. 185, no. 4, pp.

    S4-S24, 2006.

    5. V. Triggiani, F. Resta, E. Guastamacchia, C. Sabba, B. Licchelli, S. Ghiyasaldin, et al.,

    Role of antioxidants, essential fatty acids, carnitine, vitamins, phytochemicals and trace

    elements in the treatment of diabetes mellitus and its chronic complications. Endocrine,

    Metabolic and Immune Disorders Drug Targets, vol. 6, no. 1, pp. 77-93, 2006.

    6. E. Van-Quaquebeke, G. Simon, A. Andre, J. Dewelle, M. E. Yazidi, F. Bruyneel, J. Tuti, O.

    Nacoulma, P. Guissou and C. Decaestecker, Identification of a novel cardenolide (2''-

    oxovoruscharin) from Calotropis procera and the hemisynthesis of novel derivatives displaying

    potent in vitro antitumor activities and high in vivo tolerance: structure-activity relationship

    analyses. Journal of Medicinal Chemistry, vol. 48, no. 3, pp. 849-56, 2005.

    7. J.L. Hartwell, Plants Used Against Cancer, pp. 709, 1982. Lawrence, Massachusetts: This

    book lists plants reported to possess anticancer activity, covering up to 1971. It has been

    updated by Farnsworth et al. Journal of Ethnopharmacology, vol. 73, pp. 347-377, 2000.

    8. G.M. Cragg, S.A. Schepartz, M. Suffness and M.R. Grever, The taxol supply crisis. New

    NCI policies for handling the large-scale production of novel natural product anticancer and

    anti-HIV agents, Journal of Natural Products, vol. 56, no. 10, pp. 1657-1668, 1993.

    9. G.M. Cragg, M.R. Boyd, J.H. Cardellina, D.J. Newman, K.M. Snader and T.G. McCloud,

    Ethnobotany and drug discovery experience of the US National Cancer Institute. In: Chadwick,

    D.J. Marsh and J. (Eds.), Ciba Foundation Ethnobotany and the search for new drugs

    Symposium, vol. 185. Wiley and Sons, Chichester, UK, pp. 178-196, 1994.

  • 22

    10. H.K. Wang, Plant-derived anticancer agents currently in clinical use or clinical trials. The

    Investigation Drugs Journal, vol. 1, pp. 92-102, 1998.

    11. G.M. Cragg, P.G. Grothaus, and D. J. Newman, Impact of natural products on developing

    new anti-cancer agents, Chemical Reviews, vol. 109, no. 7, pp. 3012-3043. 2009.

    12. J. W. H. Li, J. C. Vederas. Drug discovery and natural products: end of an era or an

    endless frontier? , Science, vol. 325, no. 5937, pp. 161-165, 2009.

    13. A. Amin, H. Gali-Muhtasib, M. Ocker a nd R. Schneider-Stock, Overview of Major

    Classes of Plant-Derived Anticancer Drugs, International Journal of Biomedical Science, vol.

    5, no. 1, pp. 1-11, 2009.

    14. B.F. Issell, A.R. Rudolph and A.C. Louie, Etoposide (VP-16-213): an overview. In B.F.

    Issell, F.M. Muggia, and S.K. Carter, (eds.), Etoposide (VP-16-213) Current status and new

    developments, Academic Press Inc, pp. 1-13, 1984.

    15. D.C. Ayres and J.D. Loike, Lignans, chemical, biological and clinical properties,

    Cambridge University Press, 1990.

    16. M. Gordaliza, M. A. Castro, J. M. Miguel Del Corral and A. San Feliciano, Antitumor

    properties of podophyllotoxin and related compounds, Current Pharmaceutical Design, vol. 6,

    pp. 1811-39, 2000.

    17. A. Montecucco and G. Biamonti, Cellular response to etoposide treatment. Cancer

    Letters, vol. 252, no. 1, pp. 9-18, 2007.

    18. P.A. Sung, J. Libura and C. Richardson; Etoposide and illegitimate DNA double-strand

    break repair in the generation of MLL translocations, new insights and new questions. DNA

    Repair vol. 5, no. 9-10, pp. 1109-18, 2006.

  • 23

    19. G.M. Cragg and D.J. Newman, Plants as source of anticancer agents. Journal of

    Ethnopharmacology, vol. 100, pp. 72-79, 2005.

    20. K.W. Wood, W.D. Cornwell and J.R. Jackson, Past and future of the mitotic spindle as an

    oncology target, Current Opinion in Pharmacology , vol. 1, no. 4, pp. 370-7. 2001.

    21. B. Gigant, C. Wang, R.B. Ravelli, et al., Structural basis for the regulation of tubulin by

    vinblastine. Nature. vol. 435, no. 7041, pp. 519-22, 2005.

    22. M.E Wall, M.C. Wani, A.W. Nicholas , G. Manikumar, C. Tele, l. Moore , A. Truesdale, P.

    Leitner, and J.M Besterman, Plant antitumor agents: synthesis and structure activity of novel

    camptothecin analogs, Journal of Medicinal Chemistry, vol. 36, pp. 2689-700, 1993.

    23. G.S. Laco, Evaluation of Two Models for Human Topoisomerase I Interaction with

    dsDNA and Camptothecin Derivatives. Journal.pone, vol. 6, no. 8, pp. 1-7, 2011.

    24. C.J. Thomas, N.J. rahier and Hecht, Camptothecin: current perspectives, Bioorganic and

    Medicinal Chemistry, vol. 12, no. 7, pp. 1585-604, 2004.

    25. M.C. Wani, H.L. Taylor, M.E. Wall, P. Coggon and A.T. McPhail, Plant antitumor agents.

    VI. The isolation and structure of Taxol, a novel antileukemic and antitumor agent from Taxus

    brevifolia. Journal of the American Chemical Society. Soc., vol. 93, no. 9, pp. 2325-2337,

    1971.

    26. E. K. Rowinsky, L. B. Grochow, C. B. Hendricks, D. S. Ettinger, A. A. Forastiere, L. A.

    Hurowitz, W. P. McGuire, S. E. Sartorius, B. G. Lubejko, S. H. Kaufmann, and R. C.

    Donehower, Phase I and pharmacologic study of topotecan: a novel topoisomerase I

    inhibitor, Journal of Clinical Oncology, Vol. 10, no. 4, 647-656, 1992.

  • 24

    27. I. Ringel, S.B. Horwitz, "Taxol is Converted to 7-Epitaxol, a Biologically Active isomer, in

    Cell Culture Medium", Journal of Pharmacology and Experimental therapeutics, vol. 242, no.

    1, pp. 692-698, 1987.

    28. F. Teyssier, J.O. Bay, C. Dionet and P. Verrelle, Rgulation du cycle cellulaire des cellules

    exposes aux radiations ionisantes. Bulletin du Cancer. vol. 86, no. 4, pp.345-57, 1999.

    29. K. Torres and S. Band Horwitz; Mechanisms of Taxol-induced cell death are

    concentration dependant. Cancer Research, vol. 58, pp. 3620-6, 1998.

    30. T.K. Yeung, C. Germond, X. Chen and Z. Wang, The mode of action of Taxol: apoptosis

    at low concentration and necrosis at high concentration. Biochemical and Biophysical

    Research Communications, vol. 263, no. 2, pp. 398-404, 1999.

    31. D. L. Sackett and J. K. Varma. Molecular mechanism of colchicine action: induced local

    unfolding of beta-tubulin. Biochemistry, vol. 32, no. 49, pp. 13560-13565, 1993.

    32. E. Niel and J. M. Scherrmann, Colchicine today, Joint Bone Spine, vol. 73, no. 6, pp.

    672-8, 2006.

    33. I. Arnal, I. Sassoon and R. Tournebize. Dynamique du fuseau, vers une cible anti-

    cancreuse, mdecine/sciences, vol. 18, no. 12, pp. 1227-35, 2002.

    34. J. Mann, Natural products in cancer chemotherapy: past, present and future. Nature

    Reviews Cancer, vol. 2, pp. 143-148, 2002.

    35. M. S. Butler, Natural products to drugs: natural product derived compounds in clinical

    trials. Natural Product Reports, vol. 22, No. 2, pp. 162-195, 2005

    36. V. Spataro and C. Sessa, Developing new anti-cancer drugs: novel targets and

    methodological problems. Forum (Genova), vol. 9, no. 3, pp. 200-9, 1999.

  • 25

    37. S. Fulda, C. Friesen, M. Los, et al., Betulinic acid triggers CD95 (APO-1/Fas)- and p53-

    independent apoptosis via activation of caspases in neuroectodermal tumors. Cancer

    Research, vol. 57, pp. 4956-64, 1997.

    38. J. Ma, S. R. Starck, S. M. Hecht, DNA polymerase p inhibitors from Tetracera

    Boiviniana, Journal of Natural Products, Vol. 62, no. 12, pp. 1660-1663, 1999.

    39. A. Duvoix, R. Blasius, S. Delhalle, M. Schnekenburger, F. Morceau, E. Henry, M. Dicato,

    and M. Diederich, Chemopreventive and therapeutic effects of curcumin. Cancer Letters,

    vol. 223, no. 2, pp. 181-190. 2005.

    40. B.B. Aggarwal, A. Kumar, and A. C Bharti. Anticancer potential of curcumin: preclinical

    and clinical studies. Anticancer Research, vol. 23, no. 1, 363-398, 2003.

    41. T. Matsura, M. Kai, Y. Fujii, H. Ito and K. Yamada. Hydrogen peroxide-induced apoptosis

    in HL-60 cells requires caspase-3 activation. Free Radical Research, vol. 30, no. 1, pp. 73-83,

    1999.

    42. H.J. Woo, K.Y. Park and C.H. Rhu, Beta-lapachone, a quinone isolated from Tabebuia

    avellanedae, induces apoptosis in HepG2 hepatoma cell line through induction of Bax and

    activation of caspase. Journal of Medicinal Food, vol. 9, no. 2, pp. 161-168, 2006.

    43. P. Kovacic, J.R. Ames, P. Lumme, et al.; Charge transfer-oxy radical mechanism for anti-

    cancer agents. Anticancer Drug Design, vol. 1, pp. 197214, 1986. J. Ma, S. R. Starck, S. M.

    Hecht DNA polymerase p inhibitors from Tetracera Boiviniana, Journal of Natural Products,

    vol. 62, pp. 1660-1663, 1999.

    44. P. Lesca, E. Rafidinarivo, P. Lecointe, and D. Mansuy; A class of strong inhibitors of

    microsomal monooxygenases: the ellipticines. Chemico-Biological Interactions, vol. 24, pp.

    189 97, 1979.

  • 26

    45. Y.C. Kuo, P.L. Kuo, Y.L. Hsu, C.Y. Cho, and C.C. Lin, Ellipticine induces apoptosis

    through p53-dependent pathway in human hepatocellular carcinoma HepG2 cells. Life

    Sciences, vol. 78, pp. 2550-2557, 2006.

    46. C.M. Lin, H.H. Ho, G.R. Pettit, and E. Hamel, Antimitotic natural products combretastatin

    A-4 and combretastatin A-2: studies on the mechanism of their inhibition of the binding of

    colchicine to tubulin. Biochemistry, vol. 28, no. 17, pp. 69-84, 1989.

    47. G.G. Dark, S.A. Hill, V.E. Prise et al.; Combretastatin A-4, an agent that displays potent

    and selective toxicity toward tumor vasculature, Cancer Research, vol. 57, no. 10, pp. 1829-

    1834, 1997.

    48. A.J. Hayes, L.Y. Li, and M.E. Lippman, Science, medicine, and the future. Antivascular

    therapy: a new approach to cancer treatment. British Medical Journal, vol. 318, no. 7187, pp.

    853-6, 1999.

    49. A. Lansiaux and C. Bailly, Le flavopiridol, une protine kinase dpendante des cyclines.

    Bulletin du Cancer. vol 87, no 10, pp. 697-701, 2000.

    50. L.R. Kelland, Flavopiridol, the first cyclic-dependent kinase inhibitor to enter the clinic:

    current status. Expert Opin Investig Drugs. vol. 9, pp. 2903-11, 2000.

    51. S.S. Zhai, E.A. Sausville et al., Clinical pharmacology and pharmacogenetics of

    flavopiridol 1-h i.v. infusion in patients with refractory neoplasms. Anticancer Drugs vol. 14,

    no. 2, pp.125-35, 2003.

    52. M. Tan, J. Yao, and D.Yu, Over expression of the c-erbB-2 gene enhanced intrinsic

    metastasis potential in human breast cancer cells without increasing their transformation

    abilities. Cancer Research, vol. 57, pp. 1199-205. 1997.

  • 27

    53. G. Melillo, E.A. Sausville, K. Cloud, T. Lahusen, L. Varesio, and A.M. Senderowicz,

    Flavopiridol, a protein kinase inhibitor, down-regulates hypoxic induction of vascular

    endothelial growth factor expression in human monocytes. Cancer Research, 59, no. 21,

    5433-7, 1999.

    54. M. Motwani, X. Li, and G.K. Schwartz, Flavopiridol, a cyclin-dependent kinase inhibitor,

    prevents spindle inhibitor-induced endoreduplication in human cancer cells. Clinical Cancer

    Research, vol. 6, pp. 924-32, 2000.

    55. MC. Christian, JM. Pluda, TC. Ho, SG. Arbuck, AJ. Murgo, and EA. Sausville, Promising

    new agents under development by division of cancer treatment, diagnosis, and centers of the

    National Cancer Institute, Seminars in Oncology, vol. 13, no. 2, pp. 2643-2655, 1997.

    56. L. Meijer, K. Bettayeb and H. Galons, R-roscovitine (CYC202, Seliciclib). In Monographs

    on enzyme inhibitors, Volume 2. CDK inhibitors and their potential as anti-tumor agents (E.

    Yue and P.J. Smith, editors). CRC Press, Taylor & Francis, chapter 9, pp. 187-226. 2006.

    57. L. Meijer and E. Raymond, Roscovitine and other purines as kinase inhibitors. From

    starfish oocytes to clinical trials, Accounts of Chemical Research, vol. 36, no. 6, pp. 417-425,

    2003.

    58. M.M. Badawi, S.S. Handa, A.D. Kinghorn, G.A. Cordell, and N.R. Farnsworth, Plant

    anticancer agents XXVVII: antileukemic and cytotoxic constituents of Dircaoccidentalis

    (Thymelaeaceae), Journal of Pharmaceutical Sciences, vol. 72, no. 11, pp. 1285- 1287, 1983.

    59. R. Tschesche, U. Schacht, G. Legler, ber Daphnoretin, ein natrlich vorkommendes

    Derivat des 3,7-Dicumarylthers. Liebigs Annalen der Chemie, vol. 662, pp. 113-125, 1963.

    60. R. Tschesche, U. Schacht, G. Legler,ber Daphnorin, ein neues Cumaringlukosid aus

    Daphne mezereum. Naturwissenschaften, vol. 50, pp. 521-522, 1963.

  • 28

    61. R. Chakrabarti, B. Das, J. Banerji, Bis-coumarins from Edgeworthia gardneri,

    Phytochemistry, vol. 25, pp. 557-558, 1986.

    62. A. Ulubelen, B. Terem, and E. Tuzlaci, Coumarins and Flavonoids from Daphne

    gnidioides, Journal of natural products, vol. 49, no. 4, pp. 692-694, 1986.

    63. H.C. Chen., C.K. Chou, Y.H Kuo. and S.F. Yeh, Identification of a protein kinase C

    (PKC) activator, daphnoretin that suppresses hepatitis B virus gene expression in human

    hepatoma cells, Biochemical Pharmacology, vol. 52, no. 7, pp. 1025- 1032, 1996.

    64. Y.F. Liou, I.H. Hall, K.H. Lee Antitumor agents LIV: the effects of daphnoretinon in vitro

    protein synthesis of Ehrlich ascites carcinoma cells and other tissues. Journal of

    Pharmaceutical Sciences, vol. 71, no. 7, pp. 745- 749, 1982.

    65. Chuan-Li Lu, Yan-Min Li, Guo-Qiang Fu, Li Yang, Jian-Guo Jiang, Liang Zhu, Fu-Lan

    Lin, Jian Chen, Qing-Shen Lin, Extraction optimisation of Daphnoretin from root bark of

    Wikstroemia indica (L) C.A. and its anti-tumour activity tests, Food Chemistry, vol. 124, pp.

    1500-1506, 2011.

    66. C. Qing, JS. Zhang, and J. Ding, In vitro cytotoxicity of salvicine, a novel diterpenoid

    quinine, Zhongguo Yao Li Xue Bao, vol. 20, no.4, pp. 297-302, 1999.

    67. J.S. Zhang, J. Ding, Q.M. Tang, M. Li, M. Zhao, L.J. Lu, LJ Chen, and S.T. Yuan,

    Synthesis and antitumour activity of novel diterpenequinone salvicine and the analogs,

    Bioorganic & Medicinal Chemistry Letters, vol. 9, no. 18, pp. 2731-2736, 1999.

    68. Z.H. Miao, T. Tang, Y.X. Zhang, J.S. Zhang, and J. Ding, Cytotoxicity, apoptosis

    induction and downregulation of MDR-1 expression by the anti-topoisomerase II agent,

    salvicine, in multidrug-resistant tumor cells. International Journal of Cancer, vol. 106, no. 1,

    pp. 108-115, 2003.

  • 29

    69. J.Y. Lang, H. Chen, J. Zhou, Y.X. Zhang, X.W. Zhang, M.H. Li, L.P. Lin, J.S. Zhang, M.P.

    Waalkes, and J. Ding, Antimetastatic effect of salvicine on human breast cancer MDA-MB-435

    orthotopic xenograft is closely related to Rho-dependent pathway. Clinical Cancer Research,

    vol. 11, no.9, pp. 3455-3464, 2005.

    70. C.A. Austin and K.L. Marsh, Eukaryotic DNA topoisomerase II. BioEssays, vol.20, no. 3,

    pp. 215-226, 1998.

    71. Wang JC, Cellular roles of DNA topoisomerases: a molecular perspective. Nature

    Reviews Molecular Cell Biology, vol. 3, no. 6, pp. 430-440, 2002.

    72. AJ. Carpenter and AC. Porter, Construction, characterization, and complementation of a

    conditional-lethal DNA topoisomerase II alpha mutant human cell line, Molecular and

    Cellular Biology, vol. 15, no. 12, pp. 5700- 5711, 2004.

    73. Y. Pommier, M.R. Fesen, and F. Goldwasser, in Cancer Chemotherapy and Biotherapy:

    Principles and Practice , (Chabner BA and Longo DL eds), pp. 435-461, 1996. Lippincott-

    Raven Publishers, Philadelphia.

    74. TR Hammonds, A Maxwell, and JR Jenkins, Use of a rapid throughput in vivo screen to

    investigate inhibitors of eukaryotic topoisomerase II enzymes, Antimicrob Agents Chemother,

    vol. 42, no. 4, pp. 889-894, 1998.

    75. A.M. Wilstermann and N. Osheroff, Stabilization of eukaryotic topoisomerase II-DNA

    cleavage complexes, Current Topics in Medicinal Chemistry, vol. 3, no. 3, pp. 321-338, 2003.

    76. L.H. Meng, J.S. Zhang, and J. Ding, Induction of bulk and c-myc P2 promoterspecific

    DNA damage by an anti-topoisomerase II agent salvicine is an early event leading to apoptosis

    in HL-60 cells. FEBS Lett, vol. 501, no. 1, pp. 59-64, 2001.

  • 30

    77. Z.H. Miao and J. Ding, Transcription factor C-Jun activation represses Mdr-1 gene

    expression. Cancer Research, vol. 63, no. 15, pp. 4527-4532, 2003.

    78. H.R. Lu, L.H. Meng, M. Huang, H. Zhu, Z.H. Miao, and J. Ding, DNA damage, c-myc

    suppression and apoptosis induced by the novel topoisomerase II inhibitor, salvicine, in human

    breast cancer MCF-7 cells. Cancer Chemother Pharmacol vol. 55, no. 3, pp. 286-294, 2005.

    79. A. Aigner, S.S. Hsieh, C. Malerczyk, and F. Czubayko. Reversal of HER-2 over-

    expression renders human ovarian cancer cells highly resistant to taxol. Toxicology, vol. 144,

    no. (1-3), pp. 221228, 2000.

    80. K. Prabhakar, S. Ganesh, V. Baheti, S. Saisivam, Y. Madhusudan Rao and V. Kishan.

    Preparation and in vitro cytotoxic evaluation of taxol immunoconjugates. Indian journal of

    biotechnology. Vol. 6, pp 463-468, 2007.

    81. Yang Zhen-yu, Guo Wei, Wu Dong-yuan, Du Zhi-min, Study on Extraction, Isolation and

    Anti-tumor Activity of Daphnoretin from Wikstroemia indica. Natural Product Research &

    Development, vol. 20, no. 3, p522, 2004.

    82. MJ. Towle, KA. Salvato, J. Budrow, BF. Wels, G. Kuznetsov, KK. Aafls, S. Welsh, W.

    Zheng, BM. Seletsky, MH. Palme, GJ. Habgood, LA. Si,ger, LV. Dipietro, Y. Wang, JJ. Chen,

    DA. Quincy, A. Davis, K. Yoshimatsu, Y. Kishi, MJ. Yu, and BA. Littlefield, In vitro and in

    vivo anticancer activities of synthetic macro cyclicketone analogues of halichondrin B,

    Cancer Research, vol. 61, no. 3, pp. 1013-1021, 2001.

    83. LH. Zhang, L. Wu, HK. Raymon, RS. Chen, L. Corral, MA. Shirley, R K. Narla, J. Gamez,

    GW. Muller, DI. Stirling, JB. Bartlett, PH. Schafer, and F. Pawandi, The Synthetic Compound

    CC-5079 Is a Potent Inhibitor of Tubulin Polymerization and Tumor Necrosis Factor-A

    Production with Antitumor Activity, Cancer Research, vol. 66, no. 2, pp. 951-959, 2006.

  • 31

    84. BW. Parker, G. Kaur, W. Nieves-Neira, M. Taimi, G. Kohlhagen, T.Shimizu, MD.

    Losiewicz, Y. Pommier, EA. Sausville, and AM. Senderowic, Early Induction of Apoptosis in

    Hematopoietic Cell Lines After Exposure to Flavopiridol, Blood, vol. 92, no. 2, pp. 458-465,

    1998.

    85. V. Smith, F. Raynaud, P. Workman, and LR. Kelland, Characterization of a Human

    Colorectal Carcinoma Cell Line with Acquired Resistance to Flavopiridol, Molecular

    Pharmacology, vol. 60, no. 5, pp. 885893, 2001.

    86. V. Srivastava, AS. Negi, JK. Kumar, MM. Gupta and SP. Khanula, Plant-based anticancer

    molecules: A chemical and biological profile of some important leads, Bioorganic&

    Medicinal Chemistry, vol. 13, no. 21, pp. 58925908, 2005.

    87. D. Marko, S. Schtzle, A. Friedel, A. Genzlinger, H. Zankl, L. Meijer and G Eisenbrand,

    Inhibition of cyclin-dependent kinase 1 (CDK1) by indirubin derivatives in human tumour

    cells, British Journal of Cancer, vol. 84, no. 2, pp. 283289, 2001.

    88. AB. da Rocha, RM. Lopes and G. Schwartsmann, Natural products in anticancer therapy,

    Current Opinion in Pharmacology, vol. 1, no.4, pp. 364369, 2001.

    89. M. Stiborov, J. Poljakov, E. Martnkov, L. Boek-dohalsk, T. Eckschlager, R. Kizek, E.

    Frei, Ellipticine cytotoxicity to cancer cell lines a comparative study, Toxicology, vol. 4,

    no. 2, pp. 98105, 2011.

    90. ZH. Miao, T. Tang, Y-X. Zhang, J-S. Zhang, nd J. Ding, Cytotoxicity, apoptosis induction

    and downregulation of mdr-1 expression by the anti-topoisomerase ii agent, salvicine, in

    multidrug-resistant tumor cells, International Journal of Cancer , vol. 106, no. 1, pp. 108

    115, 2003.

  • 32

    91. DM. Lucas, RB. Edwards, G. Lozanski, DA. West, JD. Shin, MA. Vargo, ME. Davis, DM.

    Rozewski, AJ. Johnson, B-N. Su, VM. Goetti, NA. Heerema, TS. Lin, A. Lehman, X. Zhang,

    D. Jarjoura, DJ. Newman, JC. Byrd, AD. Kinghorn and MR. Grever, The novel plant-derived

    agent silvestrol has B-cell selective activity in chronic lymphocytic leukemia and acute

    lymphoblastic leukemia in vitro and in vivo, blood, vol. 113, no. 19, pp. 4656-4666, 2009.

    92. S-C. Chang, C-T. Huang, and S-M. Huang, Experimental Study on Effect of Compounds

    in Inhibiting HCT-116 Human Colon Cancer Cells: The Preliminary Results, Journal Society

    Colon Rectal Surgeon (Taiwan), vol. 21, pp. 121-129, 2010.

    93. Z. Huang, G. Chen, and P. Shi, Effects of emodin on the gene expression profiling of

    human breast carcinoma cells, Cancer Detection and Prevention Journal, vol. 32, no. 4, pp.

    286-29, 2009.

    94. GY. Wang, JW. Zhang, QH. Lu, RZ. Xu, and QH. Dong, Berbamine induces apoptosis in

    human hepatoma cell line SMMC7721 by loss in mitochondrial transmembrane potential and

    caspase activation, Journals of Zhejiang University-Science, vol. 8, no. 4, pp. 8(4):248-255,

    2007.

    95. YL. Wei, L. Xu, Y. Liang, XH. Xu, XY. Zhao, .Berbamine exhibits potent antitumor

    effects on imatinib-resistant CML cells in vitro and in vivo , Acta Pharmacologica Sinica, vol.

    30, no. 4, pp. 451457, 2009.

    96. D. Marcin, P. Surowiak, M. Drag-Zalesinska, M. Dietel, H. Lage and J. Oleksyszyn,

    Comparision of the Cytotoxic Effects of Birch Bark Extract, Betulin and Betulinic Acid

    Towards Human Gastric Carcinoma and Pancreatic Carcinoma Drug-sensitive and Drug-

    Resistant Cell Line, Molecules, vol. 14, no. 4, pp. 1639-1651, 2009.

  • 33

    97. Alitheen, N.B., Mashitoh, A.R., Yeap, S.K., Shuhaimi, M., Abdul Manaf, A. and Nordin,

    L. Cytotoxic effect of damnacanthal, nordamnacanthal, zerumbone and betulinic acid isolated

    from Malaysian plant sources; International Food Research Journal, vol.17, no. 3, pp. 711-

    719, 2010.

    98. S. Kim, BY. Hwang, BN. Su, H. Chai, Q. Mi, AD. Kinghorn, R. Wild, and SM. Swanson,

    Silvestrol, a potential anticancer rocaglate derivative from Aglaiafoveolata, induces apoptosis

    in LNCaP cells through the mitochondrial/apoptosome pathway without activation of

    executioner caspase-3 or -7, Anticancer Research, vol. 27, no. 4B, pp. 2175-2183, 2007.

    99. AD. Kinghorn, EJ. Carcache de Blanco, HB. Chai, J. Orjala, NR. Farnsworth, DD.

    Soejarto, NH. Oberlies, MC. Wani, DJ. Kroll, CJ. Pearce, SM. Swanson, RA. Kramer, WC.

    Rose, CR. Fairchild, GD. Vite, S. Emanuel, D. jarjoura and FO. Cope, Discovery of anticancer

    agents of diverse natural origin, Pure and Applied Chemistry, vol. 81, no. 6, pp. 1051-1063,

    2009.

  • 34

    Table I: Example of anticancer phytoactives made from herbal and their mechanisms of action

    Phytoactives Mechanism of action Plant (Family) Therapeutic use Cytotoxicity

    Lines tested IC50

    Paclitaxel

    Docetaxel

    Stop of the mitotic metaphase-

    anaphase stage: inhibits the

    depolymerization of microtubules.

    Inhibition of proliferation

    promoting cell death Taxus

    brevifolia Nutt.

    Taxus brevifolia Nutt.

    (Taxaceae)

    Taxus baccata

    (Taxaceae)

    Cancer of the ovary,

    advanced breast and lung

    [79.80]

    SK-OV-3

    MCF7

    A-549

    IC50=105 nM

    IC50= 89 nM

    IC50= 899,9 nM

    Daphnoretin Inhibition of DNA synthesis and

    protein of cancer cells

    Wikstroemia indica

    (Thymelaeaceae)

    Human hepatoma cells

    Hep3B. [81]

    AGZY-83-a

    Hep2

    HepG2

    IC50= 8.73

    IC50= 9.71

    IC50= 31.34

    Vincristine

    Vinblastine

    Vinorelbine

    Antimitotic:

    Inhibition of tubulin polymerization

    by binding to tubulin

    Catharanthus roseus

    (Nyssaceae)

    Testicular cancer,

    Hodgkin's disease and

    lymphocytic leukemia

    U937

    HL-60

    IC50= 4 nM

    IC50=2.6 nM

  • 35

    Inhibition of microtubule assembly

    Inhibition of tubulin

    polymerization.

    Inhibition of the formation and

    function of microtubules.

    Acute lung cancer and solid

    tumors [82]

    Colchicine antimitotic:

    Inhibition of tubulin

    polymerization.

    Inhibition of the formation and

    function of microtubules.

    Colchicum autumnale

    (Colchicaceae)

    Gloriosa superba L.

    (Colchicaceae)

    Leukemia and solid tumors

    [83]

    HT29

    A549

    MCF-7

    MES-SA

    IC50= 2.8 nM

    IC50= 2.0 nM

    IC50= 3.0 nM

    IC50= 2.1 nM

    Flavopiridol Cell Apoptosis

    Inhibition of angiogenesis

    Stop the cell cycle in G1 Or G2

    Amoora rohituka

    (Meliaceae)

    Dysoxylum

    binectariferum

    (Meliaceae)

    Colorectal, prostate,

    ovarian carcinoma and solid

    tumors [84,85]

    SUDHL4

    PC3

    HCT116

    SKOV3

    IC50= 120 nM

    IC50= 203 nM

    IC50= 0.042 M

    IC50=0.265 M

  • 36

    Combretastatin A-4

    Cell cycle arrest in G2 / M phase

    Destruction of specific blood

    vessels

    Inhibition of formation of new

    vessels or anti-angiogenesis

    Combretum Caffrum

    (Combretaceae)

    Colon cancer and leukemia

    [86]

    HCT-15

    L1210

    IC50= 18 nM

    IC50= 0.007M

    Indirubin Stop the cell cycle:

    Inhibition of cyclin-dependent

    kinase

    Angelica Gentiana

    Aloe Pill

    (Umbelliferae

    Lung cancer, breast and

    colorectal cancer [87]

    LXFL529L

    MCF-7

    IC50= 9.9 M

    IC50=4 M

    Camptothecin

    Topotecan, Irinotecan

    Semisynthetic derivative of

    Camptothecin

    Cell cycle arrest and cell death:

    Altered stability of topoisomerase

    I-DNA complex and inhibition of

    DNA replication

    Camptotheca

    acuminata

    (Nyssaceae)

    Leukemia and cancer of

    colon, ovarian, colorectal

    and lung [86, 88]

    L1210

    HT-29

    IC50=23 M

    IC50=0.046 M

    Podophyllotoxin

    Etoposid and Teniposid :

    Cell cycle arrest and / or apoptosis:

    Inhibition of polymerization

    Podophyllum peltatum

    (Berberidaceae)

    Cancer of lung, prostate,

    colon breast [86]

    NCI H460

    DU145

    IC50=1.1 M

    IC50=0.8 M

  • 37

    Semisynthetic derivatives

    of Podophyllotoxin

    microtubule

    Inhibition of topoisomrase II

    (double-stranded breaks in DNA,

    activation of the kinase ATM, the

    intervention of the tumor

    suppressor protein

    HT29

    MCF7

    IC50=59 M

    IC50=4.3 M

    Ellipticine

    Inhibition of topoisomerase II

    Ochrosia borbonica

    (Apocynaceae)

    Excavatia coccinea

    (Apocynaceae)

    Ochrosia elliptica

    (Apocynaceae)

    Breast cancer, leukemia,

    neuroblastoma cells and

    glioblastoma [89]

    MCF-7

    HL-60

    IMR-32

    U87MG

    IC50= 1.25 M

    IC50= 0.67 M

    IC50= 0.27 M

    IC50= 1.48 M

    Salvicine

    Inhibits the catalytic activity of

    topoisomerase II (htopo II) which

    causes DNA cleavage

    Salvia prionitis Hance

    (Lamiaceae)

    Breast cancer, leukemia and

    malignant tumors [90]

    K-562

    KB

    MCF-7

    IC50= 0.87 M

    IC50= 2.26 M

    IC50= 2.61 M

  • 38

    Silvestrol Cell apoptosis:

    mitochondrial pathway involved

    triggering the extrinsic pathway of

    programmed cell death of tumor

    cells

    Aglaia foveolata

    Panell

    (Meliaceae)

    prostate cancer, breast and

    lung cancer and leukemia

    [91,92,93]

    JeKo-1

    697 B-cell

    IC50

  • 39

    Inhibition of the enzyme involved

    in the mechanism of DNA repair,

    g/mL

    SK-OV-3 ovarian adenocarcinoma, MCF7: breast adenocarcinoma, A-549: Carcinoma of the lung, AGZY-83-a:, adenocarcinoma of the lung, HepG2:

    hepatocellular carcinoma, U937: histiocytic lymphoma, HL-60, human leukemia cells , HT29 colon Adenocarcinoma, MES-SA: Uterine sarcoma, SUDHL4:

    follicular lymphoma cells, PC3: Prostate Adenocarcinoma, HCT116: colon carcinoma, HCT-15: colon Carcinoma, L1210: animal leukemia cells (mouse),

    LXFL529L: lung carcinoma, NCI H460: carcinoma of the lung, DU145: prostate cancer, IMR-32: Neuroblastoma, U87MG: Glioblastoma, K-562: CML

    leukemia (human), KB: carcinoma of the skin, Jeko-1: acute lymphoblastic leukemia, B-697 cell: acute lymphoblastic leukemia, SMMC7721: human

    hepatoma, 181RNOV: Pancreatic carcinoma, 257RDB: Gastric carcinoma, CEM-SS: lymphocytic leukemia (human), WEHI-3B acute promyelocytic leukemia

    (human)