You are on page 1of 16

G Model

YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS


Seminars in Cancer Biology xxx (2016) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Cancer Biology


journal homepage: www.elsevier.com/locate/semcancer

Review

Potential of neem (Azadirachta indica L.) for prevention and treatment


of oncologic diseases
Shradha M. Patel a , Kalyan C. Nagulapalli Venkata b , Piyali Bhattacharyya c ,
Gautam Sethi d,e , Anupam Bishayee b,∗
a
College of Biomedical Sciences, Larkin Health Sciences Institute, Miami, FL 33169, USA
b
Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, Miami, FL 33169, USA
c
School of Health Sciences, University of Turabo, Gurabo, PR 00778, USA
d
Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
e
School of Biomedical Sciences, Curtin Health Innovation Research Institute, Biosciences Research Precinct, Curtin University, Western Australia 6009,
Australia

a r t i c l e i n f o a b s t r a c t

Article history: Throughout time, plants have often displayed medicinal properties that have been underscored. We
Received 27 November 2015 often derive medicines involved in treating cancer from components in plants. Azadirachta indica, com-
Received in revised form 19 March 2016 monly known as “neem”, has been used to treat different ailments in many Asian countries. Due to its
Accepted 21 March 2016
widespread beneficial uses, A. indica has often been referred to as “the wonder tree” or “nature’s drug
Available online xxx
store”. Various parts of this plant, including, leaves, flowers, fruits, seeds, roots, bark and oil, produce a
large number of phytochemicals with various biological and pharmacological activities. The numerous
Keywords:
biological activities of the phytoconstituents of A. indica explain its beneficial uses for the prevention
Cancer
Chemopreventive effect
and therapy of cancer. The chemopreventive and anticancer therapeutic efficacy of A. indica fractions
Therapeutic effect and compounds could be explained by multiple cellular and molecular mechanisms, including free
Phytoconstituents radical scavenging, carcinogen-detoxification, DNA repair, cell cycle alteration, programmed cell death
(apoptosis) and autophagy, immune surveillance, anti-inflammatory, anti-angiogenic, anti-invasive and
anti-metastatic activities as well as their ability to modulate several dysregulated oncogenic signaling
pathways. This article aims to present the collective and critical analysis of multiple phytoconstituents of
A. indica and their molecular mechanisms implicated in cancer chemopreventive and therapeutic effects
based on published preclinical and clinical results. Current limitations and future directions of research
on this medicinal plant are also critically discussed.
© 2016 Elsevier Ltd. All rights reserved.

1. Introduction apeutic potential, neem is also referred to as “Village pharmacy”,


“Tree of the 21st century” and “A tree for solving global problems”
Azadirachta indica (family: Meliaceae), known as neem, nimtree [1,5].
and Indian Lilac, was first discovered in India about 4500 years Today, neem tree can be found in at least 30 countries in Asia,
ago. Neem has been given the Latinized name A. indica, which is Africa, Australia as well as Central and South Americas [1]. A. indica
derived from the Persian language and literally means “the free can grow in dry and hot climates, allowing it to tolerate a tem-
tree of India” [1]. Various parts of neem tree, including leaves, flow- perature of 50–98 ◦ F [2]. A. indica has a low tolerance for rainfall
ers, fruits, seeds and bark, find extensive use in traditional systems allowing it to grow best in poor soils that are sandy, deep and have
of medicine (e.g., Ayurveda, Unani and Siddha) for treating various a pH of about 6.2–7 [2]. Usually, A. indica tree is found in dry, tropi-
human diseases, including tumor [2–4]. Due to its tremendous ther- cal or subtropical locations; however, it can also be found along the
sandy riverbanks in Australia [2].
The tree (Fig. 1A) is known to grow to approximately 15–20 m
∗ Corresponding author at: Department of Pharmaceutical Sciences, College of
high and can live for about 200 years [1]. Since the tree develops
Pharmacy, Larkin Health Sciences Institute, 18301 N. Miami Avenue, Miami, FL
a deep and strong tap root, the tree branches spread out widely
33169, USA. and form an oval crown [2]. The oil from A. indica is usually from
E-mail addresses: abishayee@ULarkin.org, abishayee@gmail.com (A. Bishayee). the seed and branches of the tree. The bark is brown and vertically
URL: http://mailto:abishayee@ULarkin.org (A. Bishayee).

http://dx.doi.org/10.1016/j.semcancer.2016.03.002
1044-579X/© 2016 Elsevier Ltd. All rights reserved.

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
G Model
YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS
2 S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx

Fig. 1. Various photographs of A. indica L. showing a whole tree (A), leaves (B), flowers (C), and fruits (D).

fissure. The leaves (Fig. 1B) are pinnate and green; however, when acids and their esters and hydrocarbons [6]. However the pres-
the plant is younger, the leaves show a purple-red color [6]. A. indica ence of phytochemicals varies in accordance with their differential
has small, drooping and fragrant white flowers (Fig. 1C) that are growth, harvesting, processing and storage conditions.
about 25 cm long each [6]. The fruit (Fig. 1D) is small, yellow and In 1942, Siddiqui [12] reported, for the first time, the isolation
edible [6]. It looks like an olive which is smooth and round with a of chemical constituents, namely nimbin, nimbinin and nimbidin,
single brown seed in the middle [6]. from neem oil. The next two decades witnessed very little success
In the last several years, neem phytoconstituents have been towards identification and isolation of any new phytochemicals.
shown to possess a plethora of biological and pharmacologi- However, the emergence of new isolation methods and improved
cal activities. These activities include and are not limited to analytical techniques for structure elucidation led to identification
anti-inflammatory, anti-pyretic, anti-histamine, anti-fungal, anti- of many important phytochemicals previously unknown [13]. The
bacterial, anti-ulcer, analgesic, anti-arrhythmic, anti-tubercular, chemical arsenal of neem is very diverse and there are reports of
anti-malarial, diuretic, spermicide, anti-arthritic, anti-protozoal, more than 300 phytochemicals isolated and characterized [14]. The
insect repellant, anti-feedant and anti-hormonal properties [1,3,7]. crude extracts prepared with water, chloroform, ethanol, butanol,
Neem-derived constituents can block cancer growth through ethyl acetate and hexane revealed the presence of numerous
diverse biomolecular and cellular mechanisms. Neem phytochem- phytochemicals. These phytochemicals are grouped based on the
icals suppress proliferation and growth of cancer cells, induce cell presence or absence of isoprene units as isoprenoids and non-
cycle arrest and apoptosis, interfere with growth factor signaling, isoprenoids (Fig. 2) [2,6].
inhibit angiogenesis, and decrease tumor cell invasion and migra- Neem isoprenoids are categorized into three classes, namely
tion. These curative powers of neem tree could be due to presence diterpenoids, triterpenoids and steroids. However, triterpenoids
of numerous chemical constituents, such as azadirachtin, gedunin, represent the major class since more than 180 triterpenoids have
nimbidin, nimbidol, nimbin, salannin and quercetin, present in been isolated from the different parts of neem tree [6,14]. Triter-
various parts of the plant. Several excellent recent reviews high- penoids have 30 carbon atoms arranged in 4-rings (A, B, C and D)
light anti-cancer pharmacological properties of A. indica [5,8–11]. with a short side chain of carbon atoms, which possess either acyclic
The purpose of this review is to collectively and critically analyze or cyclic structure. The parent triterpenoid apotirucallol (Fig. 3) has
available and up-to-date literature that presents cancer chemopre- 30 carbon atoms [6,15]. The neem triterpenoids are subdivided into
ventive and therapeutic effects of various extracts, fractions and classes based on the removal of carbon atom either from the side
phytochemicals of A. indica based on published preclinical and clin- chain or from the ring skeletal structure of the parent compound
ical results. apotirucallol [6]. The derived compounds are referred to as “nor
compounds” based on IUPAC nomenclature recommendations to
indicate the loss of carbon atom from the parent structure [16].
2. Chemical constituents of A. indica Neem triterpenoids are categorized into protolimonoids,
mononortriterpenoids, dinortriterpenoids, trinortriterpenoids,
Each anatomical part of neem tree, including the whole tree, tetranortriterpenoids, pentanortriterpenoids, hexanortriter-
stems, branches, leaves, flowers and fruits, produces different phy- penoids, octanortriterpenoids and nonanortriterpenoids.
tochemicals. The chemical composition of A. indica is very complex Protolimonoids have C-8 side chain with all the 30 carbons similar
as it contains remarkably diverse array of phytochemicals, such as to parent triterpenoid structure apotirucallol. Protolimonoids are
terpenoids, flavonoids, coumarins, carbohydrates, proteins, fatty

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
G Model
YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS
S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx 3

Fig. 2. Classification of neem chemical constituents.

considered as precursors to limonoids. The mononor, dinor, trinor, the identification and isolation of limonoids. Gualteri et al. [19] have
tetranor, pentanor, hexanor, octanor and nonanor triterpenoids recently reported the isolation of 8 limonoids from neem leaves.
are represented by the loss of 1, 2, 3, 4, 5, 6, 8 and 9 carbons, respec- Diterpenoids represent another important class of neem iso-
tively, from the parent triterpenoid apotirucallol structure. In view prenoids that are well characterized. These compounds are derived
of exhaustive literature on chemistry of neem triterpenoids, only from 4-isoprene units and their biosynthetic precursor is 20-carbon
a brief review of these compounds is given below. The interested diterpene alcohol geranylgeraniol. However, due to chemical
reader can get detailed information on neem triterpenoids from modifications occurring at later stages of biosynthetic pathway,
the existing literature [2,6,14,15,17]. numerous diterpenoids with wide variety of chemical structures
Tetranortriterpenoids, also known as limonoids, are the most are generated. There are more than 20 diterpenoids isolated from
important and well-studied class of triterpenoids and about different parts of neem tree. Neem diterpenoids mainly belong to
one-third of phytochemicals isolated from neem belong to this two classes, namely podacarpanoids (margolone) and abeitanoids
class [14]. Limonoids are oxygenated triterpenes bearing a furan (sugiol) (Fig. 3) [6,20]. Apart from triterpenoids and diterpenoids,
ring, which is formed by the loss of 4 carbon atoms from the several important steroids which are previously known, such as
side chain of the protolimonoid [6,15]. Limonoids are catego- cholesterol, b-sitosterol and stigmasterol, have also been isolated
rized into two classes based on their skeletal ring structure, from neem tree [6].
namely ring intact limonoids and ring seco limonoids. As the In addition to isoprenoids, neem tree is a rich source for num-
name suggests in ring intact limonoids, rings A, B, C and D are ber of non-isoprenoids. Various classes of non-isoprenoids include
intact, whereas in ring seco limonoids, there is a cleavage of flavonoids (quercetin, catechin and nimbaflavanone), coumarins
one or more rings of the steroidal skeleton [15,16]. Azadirone, (scopoletin), isocoumarins (margocetin), acids and their deriva-
azadiradione, isonimolide, azadirachtin, salannin, nimbolide, tives (nimbochalcin, nimbocetin, gallic acid and tiglic acid), styryl
gedunin and 7-deacetyl-7-benzoylepoxyazadiradione are few rep- derivatives (a-hexyl cinnamaldehyde and b-asarone) hydrocar-
resentative limonoids of both the classes. Azadirachtin, nimolide bons (octadecane and nonadecane), fatty acids and their derivatives
and gedunin have been extensively studied for their anti-cancer (arachidic acid, stearic acid and oleic acid), carbohydrates and pro-
properties [14]. Azadirachtin, a ring-C seco limonoid with 16 teins (D-glucose, L-arabinose and D-glucosamine), thiols (dipropyl
stereogenic centers, is the most studied limonoid and due to its sulfide) [2,6]. Structures of few selected compounds are shown in
structural complexity, it took 18 years to solve the structure. More- Fig. 4.
over, due to its unique structure and wide range of biological
properties, it was referred to as “scientific gold mine” [18]. Due to
their wide range of bioactivities, there is continued interest towards

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
G Model
YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS
4 S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx

Fig. 3. Structures of isoprenoid neem constituents.

3. A. indica and cancer eration of estrogen-dependent (MCF-7) and estrogen-independent


(MDA-231) breast carcinoma cells. Mechanistic studies showed
Research articles presented in this review exemplify the that ENLE induced cell cycle arrest at G0/G1 phase, decreased
chemopreventive and chemotherapeutic potential of A. indica B-cell lymphoma 2 (Bcl-2) and B-cell lymphoma-extra-large (Bcl-
against various cancer models. The following sections showcase xL) mRNA expression, increased B-cell associated X protein (Bax)
in vitro, in vivo and clinical studies executed around the world and Bcl-2-associated death promoter (Bad) in both breast can-
by many researchers regarding anticancer activities of neem. cer cell lines. Additionally, caspase-3 activity was increased which
Several databases, including PubMed, EBOSCOhost, and Google led to cleavage of several substrates, including poly(ADP-ribose)
Scholar, were used to find all primary literature. There were no polymerase (PARP), a nuclear enzyme involved in DNA repair
time restraints on articles that were published. The publications and maintenance. The investigators also reported downregula-
considered for this work were all in English language. First, the tion of insulin-like growth factor-1 receptor (IGF-1R), rat sarcoma
abstracts of original research papers were reviewed, followed by (Ras), Raf, p-Akt, p-Erk and cyclin D1 protein expression [22]. In
the retrieval of the full article to analyze anticancer activities of A. a separate study, the same research group treated MCF-7 and
indica. Various combinations of major keywords included: A. indica; MDA-MB-231 cells with nimbolide, a limonoid present in leaves
neem; chemopreventive; chemotherapeutic; cancer; tumor; pre- and flowers of A. indica, and observed antiproliferative activ-
vention; treatment and clinical studies. Additional relevant articles ity. The pro-apoptotic activity demonstrated by nimbolide was
were collected by studying the references of the primary articles. In mediated via the upregulation of Bax, Bad, Fas-L, tumor necrosis
order to obtain information regarding clinical studies; Clinicaltri- factor (TNF)-related apoptosis-inducing ligand (TRAIL), Fas-
als.gov was used in addition to the previously mentioned sources. associated death domain receptor (FADDR) and cytochrome c (cyt.
c) with a downregulation of Bcl-2, Bcl-xL, Mcl-1 and XIAP-1 [23].
3.1. Preclinical studies Sharma et al. [24] confirmed the cytotoxic effects of ENLE in MCF-7
cells. The treated cells exhibited apoptosis possibly through upreg-
The following sections present various anticancer studies con- ulation of Bax and downregulation of cyclin D1 and cytochrome
ducted using neem extracts and isolated pure compounds as well as P450 monooxygenases (CYP 1A1 and CYP 1A2). Collectively, these
established cancer cell lines (Table 1) and animal models (Table 2). mechanisms contributed to a decrease in cell viability and antipro-
The underlying molecular mechanisms are also analyzed. liferative activity. Several limonoid compounds isolated from A.
indica ethanolic leaf extract displayed cytotoxic activities against
SK-BR-3 breast cancer cells [25]. Desacetyl nimbinene (DAN), an
3.1.1. Breast cancer active ingredient of neem, inhibited the growth of MCF-7 as well
Crude aqueous extracts of A. indica leaves and seeds inhibited as MDA-MB-231 cells through induction of reactive oxygen species
the growth of Ehrlich ascites carcinoma cells, but no specific mech- (ROS) and loss of mitochondrial membrane potential, resulting in
anisms were identified [21]. Elumalai et al. [22] found that ethanolic mitochondria-dependent apoptotic cell death. Moreover, DAN sig-
neem leaf extract (ENLE) induced apoptosis and inhibited the prolif-

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
G Model
YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS
S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx 5

Fig. 4. Structures of steroids and non-isoprenoid neem constituents.

nificantly suppressed the migration and invasion of MDA-MB-231 Baral and Chattopadhyay [30] investigated the effect of neem
cells [26]. leaf preparation (NLP, an aqueous extract) on Ehrlich carcinoma,
Several research groups investigated breast cancer pre- a spontaneous murine mammary adenocarcinoma. Tumor-bearing
ventive and therapeutic effects of neem fractions and com- Swiss mice were treated with NLP at 1 unit/mouse/week which
ponents using various preclinical animal models. Tepsuwan led to tumor growth retardation and increased survival. Flow cyto-
et al. [27] completed a study using freeze-dried flowers of metric analysis suggested an upregulation in CD4+ and CD8+ as
A. indica in which female Sprague Dawley rats subjected to well as an increased lymphocyte count. Haque et al. [31] confirmed
9,10-dimethyl-1,2-benzanthracene (DMBA)-induced mammary that NLP restricted Ehrlich carcinoma growth in female Swiss
gland carcinogenesis were fed the test material for 20 weeks. This albino mice. The investigators prophylactically treated female
study showed a marked reduction of the incidence of mammary Swiss albino mice with NLP at 0.5, 1 and 2 units, showing an
gland tumors. Vinothini et al. [28] investigated chemopreventive increase in the production of splenic T lymphocytes. Ghosh et al.
potential of ethyl acetate and methanolic fractions of A. indica [32] investigated whether NLP could offer protection against
leaves against DMBA-induced mammary gland carcinogenesis in cyclophosphamide-induced leukopenia in normal and Ehrlich’s
female Sprague Dawley rats. Intragastric administration of both carcinoma-bearing mice. Pretreatment of mice with NLP abrogated
fractions exerted suppression of tumor incidence through inhi- the extent of leucopenia and neutropenia in normal, tumor-bearing
bition of cell proliferation, induction of apoptosis, modulation and cyclophosphamide-treated mice. Moreover, NLP pretreatment
of hormone and receptor status, alterations in xenobiotic- enhanced cyclophosphamide-mediated tumor growth inhibition
metabolizing enzymes, upregulation of antioxidant status and and host survival. An ancillary study from the same group pro-
inhibition of oxidative DNA damage. The ethyl acetate fraction vided evidence that NLP pretreatment significantly reduced the
was more effective than ethanolic fraction in modulating multiple immunotoxic effects of cisplatin and 5-fluorouracil in Ehrlich’s
molecular targets. Administration of an ethanolic fraction of neem carcinoma-bearing mice [33]. NLP has been shown to enhance Th1
leaf inhibited the growth and multiplicity of mammary tumors type immune response and anti-tumor immunity against breast
induced by N-methyl-N-nitrosourea (NMU) in female Sprague tumor-associated antigen (BTAA) in Swiss and Balb/c mice as well
Dawley rats. There was an upregulation of p53, Bax, Bad, caspases, as Sprague Dawley rats. When the intravenous injection of NLP
phosphatase and tensin homolog (PTEN) and c-Jun N-terminal was given, there was a significant increase in anti-BTAA anti-
kinase (JNK) with a downregulation of Bcl-2, angioprotein, vascular body response. Mechanistic studies showed a decrease in Th1
endothelial growth factor A (VEGF-A), cyclin D1, cyclin-dependent and interleukin-10 (IL-10) with an increase in interferon-g (IFN-
kinase 2 (Cdk2), Cdk4, and mitogen-activated protein kinase 1 g) and IgG2a antibody [34]. Othman et al. [35] evaluated the
(MAPK1) due to treatment with the neem fraction [29]. effect of ethanolic neem leaf extract on the expression of c-Myc
oncogene in xenografted 4T1 breast tumor in mice. The extract,

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
G Model
YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS
6 S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx

at 500 mg/kg, has been found to downregulate c-Myc expression in lipid peroxidation and elevation in reduced glutathione (GSH),
compared to control animals. A neem leaf glycoprotein present in glutathione peroxidase (GPx), glutathione S-transferase (GST) and
aqueous extract reduced tumor volume and increased survival mice gamma glutamyl transpeptidase (GGT) in the oral mucosa of tumor-
inoculated with Ehrlich’s carcinoma cells [36]. Banerjee et al. [37] bearing animals. Subapriya et al. [51] showed that ENLE suppressed
also used Ehrlich’s carcinoma model in which tumor growth was DMBA-induced buccal pouch carcinogenesis (squamous cell carci-
restricted and tumor angiogenesis was reduced by neem leaf gly- noma) with induction of apoptosis-related proteins Bim, caspase-3
coprotein through the downregulation of CD31, VEGF and vascular and caspase-8 as well as inhibition of Bcl-2. A follow-up study
endothelial growth factor receptor 2 (VEGFR2). from the same group [52] reported that ENLE significantly inhibited
the development of hamster buccal pouch carcinomas with simul-
3.1.2. Gastrointestinal tract and associated cancers taneous decrease in the expression of proliferating cell nuclear
Human peripheral blood mononuclear cells stimulated by NLP antigen (PCNA), mutant p53 and Bcl-2 as well as upregulation of
released IFN-g and TNF-a which triggered NLP-mediated growth cytokeratin in buccal pouch mucosa. Using the same experimen-
restriction via apoptosis in KB human oral cancer cells [38]. A tal model, Manikandan et al. [53] found that A. indica leaf fractions
neem leaf glycoprotein (NLGP) exhibited anti-tumor T cell func- (ethyl acetate and methanolic) reduced preneoplastic lesions at a
tions in KB as well as COLO205 (human colon cancer) cell lines lower concentration compared to a crude extract with simultane-
[39]. Chakraborty et al. [40] also used KB cells and found that ous modulation of phase I and phase II xenobiotic metabolizing
NLGP created an anti-tumor immune environment. NLGP inhib- enzymes, lipid and protein oxidation, upregulation of antioxidant
ited regulatory T-cell (Tregs)-induced suppression of tumoricidal defense systems, inhibition of cell proliferation and angiogenesis as
functions of CD14 + CD68 + monocyte/macrophages and inhibition well as induction of apoptosis. Priyadarsini et al. [54] investigated
of perforin/granzyme B expression. Overall, this led to the upregu- the effects of several neem phytochemicals, namely azadirachtin
lation of CD80, CD86 and HLA-ABC in monocyte/macrophages [40]. and nimbolide, on DMBA-induced buccal pouch carcinomas in
Goswami et al. [41] found that NLGP was successful in preventing male Syrian hamsters. Intragastric administration of azadirachtin
stage III supraglottic laryngeal tumor cell lysate (SLTCL)-induced and nimbolide inhibited the occurrence of DMBA-induced buccal
generation and function of pro-tumorigenic M2 tumor-associated pouch carcinomas through modulation of multiple events, includ-
macrophages. This promoted the cytotoxic activity and suppressed ing suppression of procarcinogen activation and oxidative DNA
Tregs through a downregulation of phosphorylation of targeted damage, upregulation of antioxidant and carcinogen detoxification
signal transducer and activator of transcription 3 (STAT3) in oral enzymes, and inhibition of tumor invasion and neovascularization.
squamous cell carcinoma cells. Kumar et al. [55] confirmed antitumor effects of azadirachtin and
An ethyl acetate fraction of A. indica leaf extract showed antipro- nimbolide in DMBA-induced buccal pouch carcinogenesis in male
liferative potential in HT-29 human colon adenocarcinoma cells Syrian hamsters. There was an increase in apoptosis, decrease in
[42]. Ethanolic and aqueous extracts of neem leaf were effective cell proliferation and alterations in PCNA, cyclin D1, p53, Apaf-1,
in exerting antiproliferative effects in HT-29 colon cancer cells caspase-3, GST-P, NF-kB and inhibitor of kB (IkB) following treat-
through apoptotic cell death driven by elevated production of ROS ment with neem phytochemicals. Another study was conducted
[43]. Kavitha et al. [44] showed that nimbolide, a neem-derived to evaluate chemopreventive efficacy of ethyl acetate chloroform
tetranotriterpenoid, deactivated nuclear factor-kB (NF-kB) and insoluble subfraction and methanol ethyl acetate insoluble fraction
consequently Wnt/b-catenin, a glycoprotein that controls embry- against hamster buccal pouch carcinogenesis model. The investiga-
onic development and adult homeostasis, leading to reduced cell tors concluded that there was a reduction in preneoplastic lesions
viability and elevated apoptosis in HepG2 human hepatocarci- and squamous cell carcinomas possibly due to multitargeted
noma cells. Gupta et al. [45] showed that nimbolide sensitized molecular mechanisms that involved alterations in xenobiotic
human colon cancer cells to TRAIL-mediated apoptosis via three metabolism, apoptosis induction and abrogation of NF-kB signal-
distinct mechanisms, namely ROS- and ERK-induced upregulation ing [56]. Recently, oral administration of gedunin, a neem limonoid,
of death receptor 5 (DR5) and DR4, downregulation of cell sur- was able to cease the progression of hamster buccal pouch carcino-
vival proteins, and upregulation of Bax and p53. In another colon mas by inhibiting phosphoinositide 3-kinase (PI3 K)/Akt and NF-kB
cancer cell line, WiDr, nimbolide was found to exert antiprolif- pathways via inactivation of Akt and inhibitory kB kinase (IKK),
erative effect through inhibition of cyclin A, resulting in S phase respectively. Immunoblot and molecular docking studies revealed
arrest. Nimbolide also retarded migration and invasion of tumor that suppression of the aforementioned signaling pathways may be
cells through inhibition of matalloproteinase-2 (MMP-2), MMP- mediated through inactivation of aldose reductase (AR). Gedunin
9 and VEGF as well as suppression of nuclear translocation of was also found to block neovascularization by downregulating the
p65/p50 and DNA-binding activity of NF-kB [46]. In HCT116 colon expression of miR-21 as well as VEGF and hypoxia inducible factor-
cancer cells, neem oil limonoids induced autophagy and caspase- 1a (HIF-1a) in the same tumor model [57].
dependent as well as apoptosis-inducing factor (AIF)-mediated Subapriya and Nagini [58] investigated the effects ENLE
apoptosis [47]. Recently, Yadav et al. [48] investigated the mech- in N-methyl-N’-nitro-N-nitrosoguanidine (MNNG)-induced gastric
anism of neem-induced apoptotic death in HCT116 and HT-29 carcinogenesis in male Wistar rats. The administration of ENLE
cells. Neem limonoids were able to target oxidative phosphory- led to the reduction of stomach tumor incidences and enhanced
lation (OXPHOS) system to trigger cancer cell death which did not antioxidant levels. Arivazhagan et al. [59] investigated the effect
require upregulation or activation of proapoptotic Bcl-2 family pro- of aqueous extract of neem leaf on MNNG-induced gastric car-
teins. Sastry et al. [49] reported that several analogues of nimbolide cinogenesis in male Wistar rats. This study demonstrated that the
exhibited stronger cytotoxic activities in colon cancer cell lines neem leaf extract was able to suppress cancer development and
(HT-29 and SW-620) than by the parent compound nimbolide. reverse oxidative stress in the liver by elevating GSH-dependent
Balasenthil et al. [50] investigated the effect of an aqueous antioxidants. Further chemopreventive effects were reported in
extract of neem leaves on DMBA-induced buccal pouch carcinogen- benzo(a)pyrene (B(a)P)-induced forestomach tumorigenesis in
esis model in male Syrian hamsters. There was a reduced incidence mice. There was a significant inhibition of tumor incidence as well
of oral neoplasms when the neem extract was administered at as tumor burden in B(a)P-induced forestomach papillomagenesis
100 mg/kg thrice a week for 14 weeks. The investigators showed in Swiss albino mice subjected to treatment with oral ethanolic
that antioxidants and detoxification systems were modulated due extract of A. indica leaves. There was an increase in hepatic GST,
to neem extract treatment as evidenced by a significant decrease DT-diaphorase, glutathione reductase (GR), GPx, superoxide dis-

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
G Model
YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS
S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx 7

Table 1
Antineoplastic effects of A. indica L. fractions and phytoconstituents based on in vitro studies.

Materials tested Cell lines Effects Mechanisms Conc. References

Breast cancer
Aqueous extract of leaves and seeds Ehrlich ascites Inhibited carcinoma 250–500 mg/mL Amer et al., 2010 [21]
cell growth
Ethanolic leaf extract MCF-7, MDA MB-231 Inhibited proliferation ↑Apoptosis; ⊥G0/G1; 10–50 mg/mL Elumalai et al., 2012
↓Bcl-2; ↑Bax; ↑cyt. c; [22]
↓Bcl-xL; ↑caspase-3;
↓IGF-1R; ↓Ras; ↓Raf;
↓p-Akt; ↓p-Erk; ↓cyclin D1
Nimbolide MCF-7, MDA-MB-231 Displayed ↑Apoptosis; ↑Bax, ↑Bad; 4–6 mM/mL Elumalai et al., 2012
anti-proliferative ↑Fas-L; ↑TRAIL; ↑FADDR; [23]
activity ↑cyt c; ↓Bcl-2; ↓Bcl-xL;
↓Mcl-1; ↓XIAP-1
Ethanolic leaf extract MCF-7 Showed ↑Apoptosis; ↑G0 phase; 10–500 mg/mL Sharma et al., 2014 [24]
antiproliferative ↑Bax; ↓cyclin D1; ↓CYP
activity 1A1; ↓CYP 1A2
Limonoids SK-BR-3 Exerted cytotoxicity 1–100 mM Takagi et al., 2014 [25]
Desacetyl nimbinene MCF-7, MDA-MB-231 Induced cell growth ↑Apoptosis; ↑ROS; pJNK; 10–50 mM Arumugam et al., 2015
↓p-p38 [26]

Gastrointestinal tract and associated cancer


Aqueous leaf extract KB Restricted tumor cell ↑Apoptosis; ↓Bcl-2; ↓cyclin 3 mg/200 uL Bose et al., 2007 [38]
proliferation D1; ↑caspase-3
Leaf glycoprotein KB, COLO25 Optimized anti-tumor ↑CD83; ↑CD80; ↑CD86; 1.5 mg/mL Goswami et al., 2010
T cell functions ↑CD40; ↑MHCs; ↑IL-12; [39]
↓IL-10; ↑transcription
factors; ↑ikaros; ↑CD28;
↑CD40L; ↑IFN-g; ↓IL-4
Leaf glycoprotein KB Created anti-tumor ↓Perforin/granzyme B; 1.5 mg/mL Chakraborty et al.,
immune environment ↓CTLA4; ↑CD80; ↑CD86; 2012 [40]
↑HLA-ABC
Leaf glycoprotein SCC131, SCC084 Sustained anti-tumor ↓phosphorylation of STAT3 1.5 mg/mL Goswami et al., 2014
effector functions [41]
Ethyl acetate fraction of leaf extract HT-29 Showed 650–1000 mg/mL Jafari et al., 2013 [42]
anti-proliferative
potential
Ethanolic and aqueous leaf extracts HT-29 Displayed reduced cell ↑Apoptosis; ↑ROS 0.1–0.4 mg/mL Roma et al., 2015 [43]
viability
Nimbolide HepG2 Reduced cell viability ↑Apoptosis; ↓NF-kB; 50 ng/mL Kavitha et al., 2012 [44]
↓GSK-3B; ↓b-catenin;
↓Bcl-2; ↑Bax; ↑cyt c;
↑Smac; ↑caspase-3
Nimbolide HCT116, HT-29 Sensitized cancer cells ↑ROS; ↑DR5; ↑DR4; 5 mM Gupta et al., 2011[45]
to TRAIL-induced ↓I-FLICE, ↓cIAP-1; ↓cIAP-2;
apoptosis ↓Bcl-2; ↓Bcl-xL, ↓survivin;
↑p53; ↑Bax
Nimbolide WiDr Retarded tumor cell ↑Apoptosis; ↓ERK1/2; 1.25 mM Babykutty et al., 2012
growth and migration ↑p38; ↑JNK1/2; ↓cyclin D1; [46]
↓cyclin A; ↓MMP-2;
↓MMP-9; ↓VEGF-A;
↓NF-kB
Limonoids HCT116 Induced cell death ↑Apoptosis; ↑caspase 300 mg/mL Srivastava et al., 2012
activation; ↑AIF; ↑LC3-II; [47]
↑autophagy
Liminoids HCT116, HT29 Induced cancer cell ↑Apoptosis; ↑caspase 150–300 mg/mL Yadav et al., 2016 [48]
death activation; ↓OXPHOS
Complex I; ↑ROS
Nimbolide HT-29, SW-620 Showed cytotoxic 3–103 mM Sastry et al., 2006 [49]
activity

Gynecological cancers
Nimbolide and analogs OVCAR-5 Showed 1.5–93.1 mM (IC50 ) Sastry et al., 2006 [49]
antiproliferative
activities
Nimbolide BeWo Inhibited proliferation↑Apoptosis; ↓PCNA; 0.3–3.0 mM Kumar et al., 2009 [67]
↓Bcl-2/Bax; ↑ROS;
↑Apaf-1; ↑caspase-3
Ethanolic leaf extract HeLa Exhibited cytotoxic ↑Apoptosis; ↑G0 phase; 10–500 mg/mL Sharma et al., 2014 [24]
effects ↑Bax; ↓cyclin D1; ↓CYP
1A1; ↓CYP 1A2
Azadirachtin, nimbolide HeLa Inhibited growth ↑Apoptosis; ⊥G0/G1; 2.5–200 mM Priyadarsini et al., 2010
↑p53; ↑p21; ↑Bax; [68]
↑survivin; ↓cyclin B;
↓cyclin D1; ↓PCNA; ↑ROS;
↓Bcl-2; ↓NF-kB
Neem leaf glycoprotein DCs Reduced tolerogenecity ↑2,3-Dioxygenase; ↑IDO; 1.5 mg/mL Roy et al., 2013 [69]
of DCs ↑CTLA4; ↓CD40; ↓CD83;
↓CD80; ↓CD86; ↓MHC II;
↑IL-10; ↓IL-2

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
G Model
YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS
8 S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx

Table 1 (Continued)

Materials tested Cell lines Effects Mechanisms Conc. References

Hematologic cancers
Ethanolic and aqueous leaf extracts E6-1 Decreased cell viability ↑Apoptosis; ↑ROS 0.01–0.1 mg/mL Roma et al., 2015 [43]
Aqueous leaf extract K562 Restricted tumor cell 3 mg/200 uL Bose et al., 2007 [38]
proliferation
Nimbolide U937 and HL-60, THP1 Induced 0.5–4.0 mM Roy et al., 2007 [70]
anti-proliferative
effects
Nimbolide KBM-5, U937 Suppressed cell ↑Apoptosis; ↓Bcl-2; 0.5–2.5 mM Gupta et al., 2010 [71]
proliferation ↓Bcl-xL; ↓IAP-1; ↓IAP-2;
↓cyclin D1; ↓c-Myc;
↓COX-2; ↓mmp-9; ↓VEGF;
↓ICAM-1; ↓IL-6
Liminoids HL-60 Exhibited potent ↑Apoptosis; ↑caspase-3; 2.7–3.1 mM (IC50 ) Kikuchi et al., 2011 [72]
cytotoxic activity ↑caspase-8; ↑caspase-9
Nimonol HL-60 Showed cytotoxicity ↑Apoptosis; ↑Bax; ↓Bcl-2 2.8 mM (IC50 ) Takagi et al., 2014 [25]

Lung cancer
Leaf extract and fraction A-549 Inhibited proliferation 55–56 mg/mL (IC50 ) Jafari et al. 2013 [42]
Nimbolide A-549 Decreased cell viability 15.6 mM (IC50 ) Sastry et al. 2006 [49]
Limonoids A-549 Displayed potent 7.6–19.9 mM Takagi et al., 2014 [25]
cytotoxicity

Prostate cancer
Ethanolic leaf extract PC-3 Decreased cell viability ↑Apoptosis; ↑Bax; ↓Bcl-2 10–100 mg/mL Kumar et al., 2006 [73]
Ethanolic leaf extract PC-3 and LNCaP Inhibited cell ↑Apoptosis; ↓Akt 1; ↓Akt 50–100 mg/mL Gunadharini et al.,
proliferation 2; ↓p-Akt; ↓Akt; ↓PTEN; 2011 [74]
↓PI3 K; ↓cyclin D1; ↑p21;
↑Bad; ↑cyt. c; ↑caspase-3
Ethanolic leaf extract C4-2B and PC-3M-luc2 Inhibited tumor cell ↑HMOX1; ↑AKR1C2; 5–50 mg/mL Mahapatra et al., 2011
growth ↑AKR1C3; ↑AKR1B10 [75]
Supercritical extract of leaves LNCaO-luc2 and PC-3 Suppressed tumor cell ↑Apoptosis; ↓DHT; ↓PSA; 5–25 mg/mL Wu et al., 2014 [76]
growth ↓AR; ↓integrin b1;
↓calreticulin; ↓FAK
Nimbolide and analogs PC-3 Inhibited proliferation 2.3–48.2 mM (IC50 ) Sastry et al., 2006 [49]
Limonoids LNCaP and PPC1 Induced cell death ↑Apoptosis; ↑caspase 300 mg/mL Srivastava et al., 2012
activation; ↑AIF; ↑LC3-II; [47]
↑autophagy
Nimbolide PC-3 Inhibited cell ↑Apoptosis; ↑Fas; ↑FADDR; 0.5–2.5 mM Singh et al., 2014 [77]
proliferation ↑ Bax; ↑Bad; ↑IGF-3;
↓PI3 K; ↓ Akt; ↓IGF1;
↓IGF1R; ↑cyt c; ↑caspase-8,
−9, −10, −3; ↓XIAP;
↓Bcl-2; ↓p-Akt; ↓IGF1R
Nimbolide DU145 Suppressed cell ↑Apoptosis; ↓p-STAT3; 2.5–20 mM Zhang et al., 2015 [78]
viability, invasion and ↓p-JAK1; p-JAK2; ↑ROS;
migration ↓GSH; ↑GSSG

Skin cancer
Nimbolide B16 Exhibited 0.5–5.0 mM Roy et al., 2007 [70]
antiproliferative effect
Liminoids B16 Inhibited cell viability 25 mg/mL Akihisa et al., 2009
[79], 2011 [80]
Neem leaf glycoprotein B16 Displayed cytotoxicity ↑Perforin; ↑granzyme B Barik et al., 2013 [81]

mutase (SOD) and chloramphenicol acetyltransferase (CAT), with DEN/2-AAF hepatocarcinogenesis in male Sprague-Dawley rats via
a decrease in GSH content [60]. Administration of A. indica aque- induction of apoptosis and downregulation of AFP gene.
ous leaf extract also decreased tumor burden and multiplicity in Ramzinaghara et al. [64] evaluated cancer preventive effects of
the same tumor model. Additionally, scanning electron microscopic aqueous A. indica leaf extract in dimethylhydrazine (DMH)-induced
analysis confirmed the chemopreventive action of the leaf extract colon cancer in male Wistar rats. The extract manifested a 60%
[61]. reduction in tumor incidence with a simultaneous decrease in total
Tepsuwan et al. [27] studied potential chemopreventive effect sialic acid (TSA) in the sera of experimental animals. Nimbolide,
of dietary neem flower in liver carcinogenesis in addition to mam- a type of limonoid triterpene, inhibited tumor growth in athymic
mary carcinogenesis as mentioned earlier. Dietary neem flower nu/nu mice inoculated with HCT116 colorectal cancer cells. Mech-
suppressed the incidence of benign and malignant tumors in livers anistic studies showed a decrease in the expression of proteins
of rats exposed to aflatoxin B1 (AFB1 ). There was also a substan- involved in tumor cell proliferation (cyclin D1 and c-Myc), cell sur-
tial reduction in serum GGT activity in rats that received both AFB1 vival (Bcl-2, Bcl-xL, c-IAP-1, survivin and Mcl-1), invasion (MMP-9
and neem flower compared to AFB1 control. According to a study and ICAM-1), metastasis (CXCR4) and angiogenesis (VEGF) [65].
conducted by Manal et al. [62], 5% neem leaf aqueous resulted in A polyclonal antibody generated against neem leaf glycoprotein,
complete inhibition of diethylnitrosamine (DEN)-initiated and 2- a novel immunomodulator, was found to restrict the growth of
acetylaminofluorene (2-AAF)-promoted rat hepatocarcinogenesis xenografted CT-26 colon tumor in female BALB/c mice [66]. Addi-
through downregulation of GST, GPx and a-fetoprotein (AFP). Taha tionally, aqueous neem leaf extract was effective in lowering the
et al. [63] reported that 5% A. indica aqueous leaf extract inhibited growth of implanted HT-29 colon tumors in CD1 nu/nu mice [43].

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
YSCBI-1243;
G Model
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic

Table 2
In vivo chemopreventive and anticancer effects of A. indica L. fractions and pure compounds.

Materials tested Animal models Effects Mechanisms Dose (route) Duration References

No. of Pages 16
Breast cancer
Freeze-dried neem flowers DMBA-induced Reduced tumor 10% 2 weeks Tepsuwan et al., 2002
mammary gland incidence and (diet) [27]
carcinogenesis in multiplicity
female Sprague Dawley
rats
Ethyl acetate and methanolic leaf fraction DMBA-induced Suppressed tumor ↑Apoptosis; ↑PCNA; 1–10 mg/kg 3 times/week for 12 Vinothini et al., 2009
mammary incidence ↑Bcl-2; ↓caspase-3; (p.o.) weeks [28]
carcinogenesis in ↑estradiol; ↑ER; ↓GSH;
female ↓GPx; ↓SOD; ↓CAT;
Sprague-Dawley rats ↑GST-P; ↑NF-kB;
Ethanol fraction of leaves NMU-induced Reduced tumor burden ↑p53; ↑Bax; ↑Bad; 4 mg/kg (p.o.) Every day for 4 weeks Arumugam et al., 2014
mammary and suppressed tumor ↑caspases; ↑PTEN; [29]

ARTICLE IN PRESS
S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx
carcinogenesis in progression ↑JNK; ↓Bcl-2; ↓cyclin
female Sprague Dawley D1; ↓Cdk2; ↓Cdk4;
rats ↓MAPK1
Aqueous leaf extract Ehrlich carcinoma Exerted conditional Immunomodulation 1 unit/mice/week 4 weeks Baral & Chattopadhyay,
in female Swiss mice tumor growth (p.o.) 2004 [30]
retardation
Aqueous leaf extract Ehrlich carcinoma in Restricted growth of ↑CD4+; ↑CD8+ 0.25–1.0 mg (p.o.) 1 time/week for 4 Haque et al., 2006 [31]
female Swiss mice carcinoma weeks
Aqueous leaf extract Ehrlich’s carcinoma in Inhibited tumor ↑Splenic neutrophil 1 unit 1 time/week for 4 Ghosh et al., 2006 [32]
female Swiss mice growth production; (p.o.) weeks
↓leukopenia;
↓neutropenia
Aqueous leaf extract Ehrlich carcinoma in Restricted tumor ↓Leukocyte apoptosis; 0.25 mg (p.o.) 1 time/week for 4 Ghosh et al., 2009 [33]
female Swiss mice growth Prevented ↑T cells; ↑NK cells weeks
Aqueous leaf extract Swiss mice, Balb/c mice Enhanced immune ↓Th1; ↑IFN-g; ↓IL-10; 1 unit (p.o.) 1 time/week for 4 Mandal-Ghosh et al.,
and Sprague Dawley responses to tumor ↑IgG2a weeks 2007 [34]
rats exposed to BTAA vaccines
Ethanolic leaf extract 4T1 xenograft in ↓c-Myc 250, 500 mg/kg (p.o.) Every 2 days for 4 Othman et al., 2012
female Balb/c mice weeks [35]
Leaf glycoprotein Ehrlich carcinoma Reduced tumor volume 0.25 mg 4 weeks Kundu et al., 2015 [36]
growth in female in
Swiss mice
Leaf glycoprotein Ehrlich carcinoma Restricted tumor ↓CD31; ↓VEGF; 25 mg/mice (s.c.) 1 time/week for 4 Banerjee et al., 2014
growth in female in growth and normalized ↓VEGFR2 weeks [37]
Swiss mice tumor angiogenesis

Gastrointestinal tract and associated cancers


Aqueous leaf extract DMBA-induced buccal Reduced incidence and ↓Lipid peroxidation; 100 mg/kg (p.o.) 3 times/week for 14 Balasenthil et al., 1999
pouch carcinogenesis volume of oral tumors ↑GSH; ↑GPx; ↑GST; weeks [50]
of male Syrian ↑GGT
hamsters
Ethanolic leaf extract DMBA-induce buccal Reduced tumor ↑Apoptosis; ↑Bim; 200 mg/kg (p.o.) 3 times/week for 14 Subapriya et al., 2005
pouch carcinogenesis incidence and burden ↓Bcl-2; ↑caspase-8; weeks [51], 2006 52]
in male Syrian ↑caspase-3; ↓PCNA;
hamsters ↓p53; ↑cytokeratin
Leaf fractions DMBA-induced buccal Suppressed ↓PCNA; ↓Bcl-2; 1–100 mg/kg (p.o.) 3 times/week for 14 Manikandan et al.,
pouch carcinogenesis pre-neoplastic lesions ↑caspase-3; ↑PARP; weeks 2008 [53]
in male Syrian ↓VEGF;
hamsters

9
10

YSCBI-1243;
G Model
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic

No. of Pages 16
Table 2 (Continued)

Materials tested Animal models Effects Mechanisms Dose (route) Duration References

Azadirachtin and nimbolide DMBA-induced buccal Reduced incidence of ↑GST; ↑QR; ↑SOD; 10–100 mg/kg (p.o.) 3 times/week for 14 Priyadarsini et al., 2009
pouch carcinomas in pre-neoplastic and ↑CAT; ↑GSH; ↑GPX; weeks [54]
male Syrian hamsters neoplastic lesions ↑GGT; ↑GR; ↓MMP-2;
↓MMP-9; ↓HIF-1a;
↓VEGF
Azadirachtin and nimbolide DMBA-induced buccal Reduced tumor ↓cyclin D1; ↓GST-P; 10–100 mg/kg (p.o.) 3 times/week for 14 Kumar et al., 2010 [55]
pouch carcinogenesis incidence and burden ↓PCNA; ↓NF-kB; ↑IkB; weeks
in male Syrian ↑p53; ↓Bcl-2/Bax;
hamsters ↑Apaf-1; ↑caspase-3;
↑cyt. c

ARTICLE IN PRESS
Leaf subfractions DMBA-induced buccal Reduced preneoplastic ↓PCNA; ↑Bax; ↓Bcl-2; 1–100 mg/kg 3 times/week for 14 Manikandan et al.,

S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx


pouch carcinogenesis lesions and squamous ↓NF-kB p65; ↓NF-kB (p.o.) weeks 2012 [56]
in male Syrian cell carcinomas p50; ↑IkBa; ↓IKKb;
hamsters ↓CYP1A1; ↓CYP1B1
Gedunin DMBA-induced buccal Prevented progression ↓PI3 K/Akt; ↓NF-kB; 1–10 mg/kg (p.o.) 3 times/week for 14 Kishore et al., 2015 [57]
pouch carcinomas in of tumors ↓IKK; ↓AR; ↓miR-21; weeks
male Syrian hamsters ↓VEGF; ↓HIF-1a
Ethanolic leaf extract MNNG-induced gastric Reduced incidence of ↓GSH; ↓GPx; ↓GST; 200 mg/kg (p.o.) 3 times/week for 26 Subapriya and Nagini,
carcinogenesis in male stomach tumors ↓lipid peroxidation weeks 2003 [58]
Wistar rats
Aqueous leaf extract MNNG-induced gastric Suppressed cancer ↑GSH; ↑GPX; ↑GST; 100 mg/kg (p.o.) 3 times/week for 22 Arivazhagan et al.,
carcinogenesis in male development ↑vitamin C weeks 2004 [59]
Wistar rats
Ethanolic leaf extract B(a)P-induced Inhibited tumor ↑GST; ↑DT-diaphorase; 250, 500 mg/kg (per os) Every day for 15 days Dasgupta et al., 2004
forestomach burden and reduced ↑GR; ↑GPX; ↑SOD; [60]
tumorigenesis in Swiss tumor incidence ↑CAT; ↓GSH
albino mice
Aqueous leaf extract B(a)P-induced Decreased tumor 100 mg/kg (p.o.) 3 times/week for 4 Gangar et al., 2006 [61]
forestomach burden and weeks
tumorigenesis in multiplicity
female Balb/c mice
Freeze-dried neem flowers AFB1 -induced Reduced incidence of ↓GGT 12.5% (oral) 20 weeks Tepsuwan et al., 2002
hepatocarcinogenesis tumors [27]
in male Wistar rats
Aqueous leaf extract DEN/2-AAF-mediated Repaired carcinogenic ↓AFP; ↓GST; ↓GPx 5% 10 weeks Manal et al., 2007 [62]
hepatocarconogenesis damage (drinking water)
in male Sprague
Dawley rats
Aqueous leaf extract DEN/2-AAF Reduced incidence of ↑Apoptosis; ↓AFP 5% extract 4 weeks Taha et al., 2009 [63]
hepatocarcinogenesis neoplasms (oral)
in male
Sprague-Dawley rats
Aqueous leaf extract DMH-induced colon Suppressed tumor ↓TSA 100 mg/kg (oral) 3 times/week for 20 Ramzanighara et al.,
carcinogenesis in male incidence weeks 2009 [64]
Wistar rats
YSCBI-1243;
G Model
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic

Table 2 (Continued)

No. of Pages 16
Materials tested Animal models Effects Mechanisms Dose (route) Duration References

Nimbolide HCT116 xenografts in Inhibited tumor ↓Bcl-2; ↓Bcl-xL; 5, 20 mg/kg Once daily for 10 days Gupta et al., 2013 [65]
athymic nu/nu mice growth ↓c-IAP-1; ↓survivin; (i.p.)
↓Mcl-1; ↓cyclin D1;
↓c-Myc; ↓MMP-9;
↓ICAM-1; ↓CXCR4;
↓VEGF
Neem leaf glycoprotein CT-26 colon carcinoma Restricted tumor Immunomodulation 25 mg (s.c.) 1 time/week for 4 Das et al., 2014 [66]
in athymic nude mice growth weeks
Aqueous leaf extracts HT-29 xenografts in Inhibited tumor 450 mg/kg/day 47 days Roma et al., 2015 [43]
CD-1 nu/nu mice growth (d.w.)

Prostate cancer

ARTICLE IN PRESS
Ethanolic leaf extract C4-2B and PC-3M-luc2 Suppressed tumor 100, 200 mg/kg 6 times/week; Mahapatra et al., 2011

S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx


↑Apoptosis
xenografts in nu/nu growth (i.p.) 8–11 weeks [75]
mice
Supracritical leaf extract LNCaP-luc2 xenografts Retarded tumor ↓PSA; ↓AKR1C2 100, 200 mg/kg 6 times/week;9 weeks Wu et al., 2014 [76]
in nu/nu mice growth (p.o.)
Nimbolide TRAMP Inhibited tumor ↑Apoptosis; ↓Ki-67; 3 mg/kg 5 times/week; 6–12 Zhang et al., 2015 [78]
growth and metastasis ↓p-STAT (p.o.) weeks

Skin cancer
Ethanolic leaf extract DMBA-induced skin Inhibited tumor ↑GST; ↑DT-diaphorase; 250, 500 mg/kg (p.o.) Every day for 15 days Dasgupta et al., 2004
papillomagenesis in burden and reduced ↑GR; ↑GPX; ↑SOD; [60]
Swiss albino mice tumor incidence ↑CAT; ↓GSH
Aqueous leaf extract DMBA/TPA-induced Reduced mean tumor ↑Bax; ↓Bcl-2; ↑caspase 300 mg/kg (p.o.) 2–3 times/week for 20 Arora et al., 2011 [82]
skin carcinogenesis in burden and mean 3; ↑caspase 9; weeks
male LACA mice tumor volume
Aqueous leaf extract DMBA/TPA-induced Showed ↑Lipid peroxidation; 300 mg/kg (p.o.) 2 times/week for 20 Arora et al., 2013 [83]
skin carcinogenesis in anti-neoplastic activity ↓PCNA; ↑mdm2; ↑p53; weeks
male LACA mice and regulated cell cycle ↑p21
Azadirachtin ONOO− -initiated Reduced incidence and 780 nmol 2 weeks Akihisa et al., 2009 [79]
TPA-promoted skin multiplicity of
carcinogenesis in mice papillomas
Aqueous leaf extract B16 melanoma tumor Exerted tumor growth Immunomodulation 1 unit/mice/week 4 weeks Baral and
in C57BL/6 mice retardation (p.o.) Chattopadhyay, 2004
[30]
Leaf glycoprotein B16F10 melanoma Restricted tumor ↓CD31; ↓VEGF; 25 mg/mice (s.c.) 1 time/week for 4 Banerjee et al., 2014
tumor in C57BL/6 mice growth and normalized ↓VEGFR2 weeks [37]
tumor angiogenesis

Connective tissue cancers


Leaf glycoprotein Sarcoma 180 in female Restricted sarcoma ↓GATA3; ↑IFN-g 25 mg/mouse 1 time/week for 4 Mallick et al., 2013 [84]
Swiss mice growth (s.c.) weeks
Leaf glycoprotein Sarcoma 180 in female Restricted sarcoma ↑IL-2; ↑IL-12; ↑IFN-g; 25 mg/mouse 1 time/week for 4 Barik et al., 2013 [85]
Swiss mice growth ↓IL-6; ↓IL-10; ↓TGF-b (s.c.) weeks
Leaf glycoprotein Sarcoma 180 in female Reduced tumor volume ↑Perforin; ↑granzyme; 2 × 105 1 time/week for 4 Mallick et al., 2014 [86]
Swiss mice ↑IFN-g; ↑antigen cells/100 ml/mouse weeks
specific T-cell
proliferation

11
G Model
YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS
12 S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx

3.1.3. Gynecologic cancers 3.1.6. Prostate cancer


Nimbolide and its analogs showed cytotoxic activities against Kumar et al. [73] have found that an ethanolic extract of A. indica
OVCAR-5 ovary cancer cells [49]. Kumar et al. [67] found that nim- leaves decreased cell viability in PC-3 prostate cancer cells by induc-
bolide treatment resulted in dose- and time-dependent inhibition ing apoptosis. There was an increase in Bax and decrease in Bcl-2
of growth of human choricarcinoma (BeWo) cells. Mitochondria- protein. Gunadharini et al. [74] made similar findings in prostate
mediated apoptosis in BeWo cells was due to the decrease in cancer cell lines, PC-3 and LNCaP, in which ENLE inhibited cell pro-
Bcl-2/Bax ratio with an increase in Apaf-1 and caspase-3 and the liferation and induced apoptosis with simultaneous decrease in
cleavage of PARP. Treatment of HeLa cervical cancer cells with ENLE cyclin D1 and increase in p21, cyt. c and caspase-3. In another study,
retarded the growth of these cells in a dose- and time-dependent Mahapatra et al. [75] found that ethanol extract of neem leaves
manner via apoptosis induction. ENLE also differentially modu- (EENL) inhibited the growth of prostate cancer cell lines, namely
lated the expression of Bax, cyclin D1, CYP 1A1 and CYP 1A2 [24]. C4-2B and PC-3M-luc2. Microarray analysis revealed differential
Priyadarsini et al. [68] showed that azadirachtin and nimbolide regulation of various genes. Most of the genes that were upregu-
suppressed the viability of HeLa cervical cancer cells. The same lated were associated with cell death and drug metabolism while
researchers also observed that an increase in apoptosis in HeLa cells genes associated with cell cycle regulation, DNA replication, recom-
was accompanied by a blockade in G0/G1 phase, increase in p53, bination and repair functions were downregulated. Additionally,
p21, ROS, Bax and survivin as well as decrease in cyclin B, cyclin D, the quantitative PCR analysis revealed increase in gene expres-
PCNA, Bcl-2 and NF-kB. In another study, Roy et al. [69] showed that sion levels of HMOX1, AKR1C2, AKR1C3 and ARK1B10. Overall, this
neem leaf glycoprotein inhibited 2,3 dioxygenase (IDO) induction study showed that the pleiotropic effect of EENL was regulated
in co-culture of dendritic cells (DCs) and regulatory T (Tregs) cells through multiple cellular pathways in prostate cancer. Wu et al.
obtained from patients with cervical cancer stage IIIb (CaCx-IIIB). [76] studied antitumor effect of a supracritical extract of neem
Specifically, the cytotoxic T-lymphocyte antigen 4 (CTLA4) induced leaves (SENL) using LNCaP-luc2 and PC-3 prostate cancer cells.
optimal amount of DC maturation, decreased CD40, CD83, CD80, Treatment of both cells lines with SENL suppressed cell growth,
CD86, MHC-II and IL-2 with an increased IL-10. The investigators induced apoptosis and inhibited dihydrotesterone (DHT)-induced
were able to conclude that there was a reduced tolerogenecity of androgen receptor (AR) and prostate-specific antigen (PSA) levels.
DCs. Moreover, SENL inhibited integrin b1, calreticulin and focal adhesin
kinase (FAK) activation. Sastry et al. [49] have found that nimbolide
and its analogs possessed cytotoxic activities against PC-3 prostate
cancer cells. Neem oil limonoids induced p-53-independent apo-
3.1.4. Hematological cancer
ptosis and autophagy in LNCap and PPC1 cells [47]. Singh et al. [77]
Both ethanolic and aqueous extracts of neem leaf reduced
confirmed that nimbolide inhibited the proliferation of PC-3 cells
the viability of E6-1 leukemic cells by induction of apoptosis
by inducing apoptosis. Mechanistic studies exemplified an increase
through destabilization of cancer cell mitochondria without harm-
in mRNA levels of Fas ligand, Fas-associated death domain recep-
ing normal cells [43]. Supernatant from human peripheral blood
tor (FADDR), Bax, Bad and insulin growth factor-binding protein 3
mononuclear cells stimulated by a neem leaf preparation restricted
(IGF-3), increased protein expressions of cyt. c, caspase-8, caspase-
the proliferation of K562 erythroleukemic cells [38]. Nimbolide was
9, caspase-10 and caspase-3 and decreased protein levels of PI3 K,
found to exhibit antiproliferative activities against several leukemic
Akt, IGF1, IGF1R, XIAP and Bcl-2. Nimbolide inhibited cell viability,
cell lines, such as HL-60, U937 and THP1. In U937 cells, nimbolide
induced apoptosis and suppressed cellular invasion and migration
treatment also resulted in cell cycle disruption by decreasing num-
of DU145 and LNCaP cells. Moreover, nimbolide abrogated STAT3
ber of cells in G0/G1 phase with an initial increase in S and G2/M
activation in DU145 cells and this effect was found to be mediated
phases. When the cells were exposed to nimbolide for a longer
via an increased production of ROS due to GSH/oxidized glutathione
time, there was an increase in damaged DNA and consequently a
(GSSG) imbalance [78].
substantial increase in the numbers of cells in sub-G1 fraction and
There are only few reports on in vivo anti-prostate cancer
decrease of cells in all phases [70]. In a separate study, nimbolide
effects of neem. In one study, EENL inhibited the growth of
suppressed the proliferation of KBM-5 (human chronic myeloid
xenografted C4-2B and PC-3M-luc2 prostate tumors in mice. The
leukemia) as well as U937 cells through proapoptotic mechanisms.
tumor inhibitory effects were associated with the formation of
Nimbolide also abrogated the expression of proteins associated
hyalinized fibrous tumor tissue and induction of apoptosis [75]. In
with proliferation (cyclin D1), survival (Bcl-2, Bcl-xL, IAP-1 and
another study, oral administration of SENL inhibited LNCaP-luc2
IAP-2), invasion (MMP-9) and angiogenesis (VEGF) by deactivating
xenograft tumor growth in mice. Accompanying immunohisto-
NF-kB pathway [71]. Kikuchi and coworkers [72] have reported
chemical analyses revealed that there was a simultaneous decrease
the cytotoxic activities of several limonoids, namely 7-deacetyl-
in PSA and increase in aldo-keto reductase family 1 member C2
7-benzoyl-epoxyazadiradione, 7-deacetyl-7-benzoylgedunin and
(AKR1C2) expression [76]. Recently, oral administration of nim-
28-deoxonimbolide, against HL60 leukemia cells via both the
bolide suppressed tumor growth and metastasis (to lungs and
mitochondrial- and death receptor-mediated pathways. Takagi
liver) in transgenic adenocarcinoma of mouse prostate (TRAMP)
et al. [25] also found that nimonol, a neem limonoid, displayed
model. The investigators also observed that nimbolide decreased
cytotoxic activities against HL-60 cells mainly due to apoptosis
the expression of Ki-67, increased cleaved-caspase-3 and down-
induction.
modulated STAT3 signaling in TRAMP tumor tissues [78].

3.1.5. Lung cancer 3.1.7. Skin cancer


While crude leaf extract and its ethyl acetate fraction showed The B16 murine melanoma cell culture model was used to
modest antiproliferative effects against A-549 lung cancer cells investigate anticancer effects of various neem extracts and phyto-
[42], nimbolide exhibited potent cytotoxic activity against the chemicals. Nimbolide treatment (0.5–5.0 mM) resulted in moderate
same cancer cell line [49]. Takagi et al. [25] reported that several to strong growth inhibition of B16 cells [70]. Several limonoids iso-
limonoids (e.g., nimonol. isonimocinolide and 28-deoxonimbolide) lated from the seed extract of A. indica exhibited cytotoxicity as
isolated from A. indica leaf showed potent cytotoxic activity against well as anti-inflammatory activities [79,80]. CD8+ T cells exposed to
A-549 cells. neem leaf glycoprotein inflicted cytotoxicity to B16 cells in vitro and

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
G Model
YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS
S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx 13

these tumor cells exhibited high expressions of cytotoxicity-related 4. Toxicity studies


molecules, such as perforin and granzyme B [81].
Dasgupta et al. [60] evaluated the chemopreventive poten- Given that neem-based preparations have been consumed for
tial of neem leaf in DMBA-induced skin papillomagenesis in several millennia, the normal consumption of neem products can
Swiss albino mice. Oral feeding of an ethanolic leaf extract be regarded as absolutely safe [2]. However, the question arises
reduced tumor burden and tumor incidence. The extract inhib- whether this safety extends to extracts or pure compounds that
ited chemical carcinogenesis through elevation of antioxidant may be used in a more concentrated form for treating or pre-
enzymes as well as modulation of phase II detoxification enzymes. venting different diseases, including cancer. Toxicological studies
Arora et al. [82] studied DMBA/tetradecanoylphorbol-13-acetate suggested that the differences in the solvents and methods used
(TPA)-induced skin carcinogenesis in male LACA mice that were to prepare the extracts could affect the toxicity level [89]. In mice,
administered 300 mg/kg of aqueous leaf extract 2–3 times a week the LD50 value of methanolic leaf extract has been shown to be
for 20 weeks. The investigators observed that the mean tumor 13 g/kg body weight [90]. Recently, Kurpadinum et al. [91] have
burden and mean tumor volume were reduced with concomitant reported similar LD50 value (12 g/kg body weight) of methanolic
induction of apoptosis. There was an increase in Bax, caspase- extract of neem flowers in rats. Dorababu and co-workers [92] have
3 and caspase-9, while decrease in Bcl-2. The same group [83] found that the aqueous extract of neem leaf is non-toxic to mice
later performed mechanistic studies on animals that were treated and they reported LD50 as >2.5 g/kg body weight. The acute oral
in the same manner, as in the previous study, and found low toxicity in rats fed with technical grade azadirachtin in a single
expression of PCNA and cyclin D1 as well as high expression of dose was greater than 5 g/kg both in male and female rats [93].
p53 and p21 in tumors obtained from neem extract-treated mice Tarbousch and Ashmaoui [94] have demonstrated that oral doses
compared to carcinogen control animals. Azadirachtin B elicited of azadirachtin have not produced any skeletal or morphological
chemopreventive effects against a two stage carcinogenesis model changes on fetuses and pups in mice. Srivastava and Raizada [95]
of mouse skin tumor induced by peroxynitrite (ONOO− ) as an ini- demonstrated that there were no adverse effects on the reproduc-
tiator and TPA as a promoter [79]. An aqueous extract of neem leaf tive function or fetal development in rats fed diets containing the
showed significant reduction of B16 melanoma tumor growth in equivalent of 5, 25 and 50 mg/kg of azadirachtin daily. However, no
mice with increased survivability possibly due to neem-mediated information on the safety on neem leaf during lactation was iden-
immune activation [30]. A neem leaf glycoprotein restricted B16F10 tified. Azadirachtin is registered in the United States as a general
melanoma growth in mice through anti-angiogenic effect [37]. use pesticide with a toxicity classification of IV (relatively non-
toxic) [96]. Neem seed oil was found to be non-toxic to rats and
rabbits when given via intragastric route. However, when given
intravenously or intraperitoneally, it caused death in rats and rab-
3.1.8. Connective tissue cancers bits with 24 h LD50 values of 14 mL/kg and 24 mL/kg, respectively
Mallick et al. [84] used therapeutic potential of NLGP against sar- [97]. Gandhi et al. [98] reported similar toxic effects in these two
coma growth in female Swiss mice. Tumor-bearing animals were species. In humans, the non-aqueous extracts have shown the pres-
treated with NLGP subcutaneously at 25 mg/mouse/injection once ence of allergens in the skin prick test for allergenic activity [99].
a week for 4 weeks. NLGP was able to restrict sarcoma growth Pure azadirachtin at a daily dose of 15 mg/kg body weight showed
in experimental animals. Mechanistically, this therapy recruited low toxicity in humans [89].
type-1 antitumor CD8+ T cells into the tumor environment and
reduced immunologic indices associated with immune suppres-
sion. A follow-up study conducted by the same research group
revealed NLGP-mediated upregulation of IL-2, IL-12 and IFN-g 5. Conclusion and future directions
with a downregulation of IL-6, IL-10 and TGF-b restricted murine
sarcoma growth [85]. An extended study by Mallick et al. [86] wit- From the studies highlighted in this review, it is apparent that
nessed a restriction in murine sarcoma growth with NLGP-matured the ethnomedicinal plant, A. indica, has the potential to prevent
sarcoma antigen-pulsed dendritic cells (DCNLGPTAg). DCNLGPTag and treat a wide array of human cancers. The chemopreventive and
activated CD8+ T cells to induce tumor cell killing and increased anticancer therapeutic efficacy of A. indica fractions and compounds
perforin, granzyme B and IFN-g secretion. could be explained by multiple cellular and molecular mecha-
nisms, including free radical scavenging, carcinogen-detoxification,
DNA repair, cell cycle alteration, programmed cell death (apo-
ptosis) and autophagy, immune surveillance, anti-inflammatory,
3.2. Clinical studies anti-angiogenic, anti-invasive and anti-metastatic activities as well
as their ability to modulate several dysregulated oncogenic signal-
Vasenwala et al. [87] conducted a clinical study to determine the ing pathways.
ability of neem extract to induce apoptosis in cervical cancer cells A careful examination of in vitro and in vivo studies presented
as well as to estimate caspase activity and TNF-a and IFN-g levels here shows that A. indica-derived constituents are effective in pre-
in monocytes from cervical cancer patients. Neem-treated mono- venting or treating cancers of the following organs: breast, cervix,
cytes from the cervical cancer patients displayed high activity levels colon, intestines, liver, lung, oral cavity, ovary, prostate, skin, stom-
of capase-3, caspase-8 and caspase-9. There was an increase in ach and uterus. However, further studies need to be conducted to
IFN-g and decrease in TNF-a level in culture supernatant of mono- specify how the mechanisms work to treat each cancer type. A
cytes. Additionally, cyto- and histo-morphology of neem-exposed majority of studies showing potential for cancer prevention and
cervical cancer cells exhibited increased apoptosis. Franco et al. treatment utilized neem leaves. Other parts of the plant, such as
[88] reported that neem oil (Holoil® RIMOS srl, Mirandola, Italy) the stem, flowers, fruits, and seeds, may also represent valuable
reduced acute skin toxicity in head and neck cancer patients who sources for anti-cancer drug development. However, a high level of
were undergoing radiotherapy or chemo-radiotherapy. The inves- quality control measures and improved standards for production
tigators indicated a prophylactic effect of the neem product in the of neem-derived phytochemicals are necessary considering these
prevention of moist desquamation in this single-arm prospective components could be greatly different due to various factors in cul-
observational study. tivation (e.g., soil and climate) and extraction process (e.g., different

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
G Model
YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS
14 S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx

solvents) as well as raw material used (leaves, flowers, fruits, seeds [7] R. Tiwari, A.K. Verma, S. Chakraborty, K. Dharma, S.V. Singh, Neem
and bark). (Azadirachta indica) and its potential for safeguarding health of animals and
humans: a review, J. Biol. Sci. (2014) 110–123.
It is not possible to precisely identify active compounds respon- [8] F. Hao, S. Kumar, N. Yadav, D. Chandra, Neem components as potential
sible for anticancer activities of A. indica. Based on studies presented agents for cancer prevention and treatment, Biochim. Biophys. Acta 1846
here, nimbolide and azadirachtin are two bioactive neem com- (2014) 247–257.
[9] L.N. Bodduluru, E.R. Kasala, N. Thota, C.C. Barua, R. Sistla, Chemopreventive
pounds studied extensively. However, as mentioned earlier, A. and therapeutic effects of nimbolide in cancer: the underlying mechanisms,
indica has numerous phytoconstituents which include alkaloids, Toxicol. In Vitro 28 (2014) 1026–1035.
carbohydrates, tannins, reducing sugars, flavonoids, glycoside, phe- [10] P. Elumalai, J. Arunakaran, Review on molecular and chemopreventive
potential of nimbolide in cancer, Genom. Inform. 12 (2014) 156–164.
nolic compounds and saponins along with amino acids aspartic
[11] S. Nagini, R.V. Priyadarsini, Azadirachta indica (neem) and neem limonoids
acid, asparagine, glutamine, serine, histidine, alanine, proline, as anticancer agents: molecular mechanisms and targets, in: S. Perumana, V.
tyrosine, methionine, cysteine, isoleucine, leucine, phenylalanine Oommen, M.R. Pillai (Eds.), Perspectives in Cancer Prevention-Translational
Cancer Research, Springer, India, New Delhi, 2014, pp. pp. 45–60.
and lysine. Even though several of these components have been
[12] S. Siddiqui, A note on the isolation of three bitter principles from the neem
acknowledged for observed beneficial effects, the proficiency of oil, Curr. Sci. 11 (1942) 278–279.
each phytochemical should be further investigated. It is likely that [13] T.R. Govindachari, Chemical and biological investigations on Azadirachta
neem uses the pleiotropic effects of various phytoconstituents to indica (the neem tree), Curr. Sci. 63 (1992) 117–122.
[14] A. Akhila, K. Rani, Chemistry of the neem tree (Azadirachta indica A. Juss),
exert broad chemopreventive and anti-cancer activities. Emerg- Prog. Chem. Org. Nat. Prod. 78 (1999) 47–149.
ing evidence strongly suggests that specific mechanisms need to [15] Q.G. Tan, X.D. Luo, Meliaceous limonoids: chemistry and biological
be studied to determine the best neem phytochemical constituent activities, Chem. Rev. 111 (2011) 7437–7522.
[16] P.M. Giles, Revised section F: natural products and related compounds, Pure
required to prevent and treat cancer. It is also possible that the com- Appl. Chem. 71 (1999) 587–643.
bination of varying constituents, or phytochemical synergy, may be [17] S. Siddiqui, B.S. Siddiqui, S. Faizi, T. Mahmood, Tetracyclic triterpenoids and
responsible for suppression of tumor development or inhibition of their derivatives from Azadirachta indica, J. Nat. Prod. 51 (1988) 30–43.
[18] D.E. Morgan, Azadirachtin a scientific gold mine, Biorg. Med. Chem. 17
tumor cell growth. (2009) 4096–5105.
Although there have been an impressive number of preclini- [19] M.J. Gualtieri, N. Malafronte, A. Vassallo, A. Braca, R. Cotugno, M. Vasaturo,
cal studies performed on A. indica as a therapeutic or preventative N.D. Tommasi, F.D. Piaz, Bioactive limonoids from the leaves of Azadirachta
indica (Neem), J. Nat. Prod. 77 (2014) 596–602.
plant against cancer, the lack of human clinical trials precludes [20] I. Ara, B.S. Siddiqui, S. Faizi, S. Siddiqui, Structurally novel diterpenoid
clinical drug development from this medicinal and dietary plant. constituents from the stem bark of Azadirachta indica (Meliaceae), J. Chem.
Randomized-controlled trials are urgently required so that the Soc. Perkin Trans. 1 (1989) 343–345.
[21] H. Amer, W.A. Helmy, H.A.A. Tale, In vitro antitumor and antiviral activities
preclinical evidence highlighted can relay clinical success in the
of seeds and leaves of neem (Azadirachta indica) extracts, Int. J. Acad. Res. 2
future. Another area for future research could be the develop- (2010) 47–52.
ment of drugs using advanced technology such as nanoformulation [22] P. Elumalai, D.N. Gunadharini, K. Senthilkumar, S. Banudevi, R. Arunakumar,
and liposomal drug delivery to improve the efficacy of neem- C.S. Benson, G. Sharmila, J. Arunakaran, Ethanolic neem (Azadirachta indica
A. Juss) leaf extract induces apoptosis and inhibits the IGF signaling pathway
based therapeutic treatments. However, possible adverse effects in breast cancer cell lines, Biomed. Prev. Nutr. 2 (2012) 59–68.
of neem constituents need to be taken into consideration. Cur- [23] P. Elumalai, D.N. Gunadharini, K. Senthilkumar, S. Banudevi, R. Arunakumar,
rently, very limited information is available on pharmacokinetic C.S. Benson, G. Sharmila, J. Arunakaran, Induction of apoptosis in human
breast cancer cells by nimbolide through extrinsic and intrinsic pathway,
profiles (absorption, distribution, metabolism and excretion) of Toxicol. Lett. 215 (2012) 131–142.
neem components. It is very critical for neem-based anti-cancer [24] C. Sharma, A.J. Vas, P. Goala, T.M. Gheewala, T.A. Rizvi, A. Hussain, Ethanolic
drug development and more studies are required in this area. Since neem (Azadirachta indica) leaf extract prevents growth of MCF-7 and HeLa
cells and potentiates the therapeutic index of cisplatin, J. Oncol. 2014 (2014)
at least half of cancer cases and mortality are known to occur in low- 321754.
and middle-income countries [100], exploring lower-cost alterna- [25] M. Takagi, Y. Tachi, J. Zhang, T. Shinozaki, K. Ishii, T. Kikuchi, M. Ukiya, N.
tives to high-cost therapies, such as development of neem-based Banno, H. Tokuda, T. Akihisa, Cytotoxic and melanogenesis-inhibitory
activities of limonoids from the leaves of Azadirachta indica (neem), Chem.
cancer preventive and therapeutic drug, would have an enormous
Biodivers. 11 (2014) 451–468.
global impact. [26] A. Arumugam, R. Subramani, S. Nandy, S. Powell, M. Velazquez, A. Orozco, A.
In conclusion, while A. indica shows significant promise for pre- Galvez, R. Lakshmanaswamy, Desacetyl nimbinene inhibits breast cancer
growth and metastasis through reactive oxygen species mediated
venting and treating cancer, more studies need to be conducted on
mechanisms, Tumor Biol. (2015) (Dec 4. [Epub ahead of print] PMID:
A. indica-based products as efficacious drugs. The preclinical stud- 26637227).
ies presented and analyzed in this review highly suggest that A. [27] A. Tepsuwan, P. Kupradinun, W.R. Kusamran, Chemopreventive potential of
indica or the “wonder tree” has a grand ability to prevent and treat neem flowers on carcinogen-induced rat mammary and liver carcinogensis,
Asian Pac. J. Cancer Prev. 3 (2002) 231–238.
human cancers. [28] G. Vinothini, P. Manikandan, R. Anandan, S. Nagini, Chemoprevention of rat
mammary carcinogenesis by Azadirachta indica leaf fraction: modulation of
hormone status, xenobiotic-metabolizing enzymes, oxidative stress, cell
6. Conflict of interest proliferation and apoptosis, Food Chem. Toxicol. 47 (2009) 1852–1863.
[29] A. Arumugam, P. Agullo, T. Boopalan, S. Nandy, R. Lopez, C. Gutierrez, M.
Authors disclose no financial conflicts of interest. Narayan, L. Rajkumar, Neem leaf extract inhibits mammary carcinogenesis
by altering cell proliferation, apoptosis, and angiogenesis, Cancer Biol. Ther.
15 (2014) 26–34.
References [30] R. Baral, U. Chattopadhyay, Neem (Azadirachta indica) leaf mediated
immune activation causes prophylactic growth inhibition of murine Ehrlich
[1] V.S. Kumar, V. Navratnam, Neem (Azadirachta indica): Prehistory to carcinoma and B16 melanoma, Int. Immunopharmacol. 4 (2004) 355–366.
contemporary medicinal uses to humankind, Asian Pac. J. Trop. Biomed. 3 [31] E. Haque, I. Mandal, S. Pal, R. Baral, Prophylactic dose of neem (Azadirachta
(2013) 505–514. indica) leaf preparation restricting murine tumor growth is nontoxic,
[2] H.S. Puri, Neem The Divine Tree Azadirachta Indica Amsterdam, Harwood hematostimulatory and immunstimulatory, Immunopharmacol.
Academic Publishers, Australia, The Netherlands, 1999. Immunotoxicol. 28 (2006) 33–50.
[3] K. Biswas, I. Chattopadhyay, R.K. Banerjeee, U. Bandyopadhay, Biological [32] D. Ghosh, A. Bose, E. Haque, R. Baral, Pretreatment with neem (Azadirachta
activities and medicinal properties of neem (Azadirachta indica), Curr. Sci. 82 indica) leaf preparation in swiss mice diminishes leukopenia and enhances
(2002) 1136–1345. the antitumor activity of cyclophosphamide, Phytother. Res. 20 (2006)
[4] S. Drabu, S. Khatri, Babu S. Neem, Healer of all ailments, Res. J. Pharm. Biol. 814–818.
Chem. Sci. 3 (2012) 120–126. [33] D. Ghosh, A. Bose, E. Haque, R. Baral, Neem (Azadirachta indica) leaf
[5] R. Paul, M. Prasad, N.K. Sah, Anticancer biology of Azadirachta indica l preparation prevent leukocyte apoptosis mediated by cisplatin plus
(neem), Cancer Biol. Ther. 12 (2011) 467–476. 5-fluorouracil treatment in Swiss mice, Chemotherapy 55 (2009) 137–144.
[6] K.K. Singh, Neem, a Treatise, IK International Publishing House Pvt. Ltd.,
New Delhi, India, 2008.

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
G Model
YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS
S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx 15

[34] I. Mandal-Ghosh, U. Chattopadhyay, R. Baral, Neem leaf preparation [56] P. Manikandan, S.M. Ramalingam, G. Vinothini, V.P. Ramamurthi, I.P. Singh,
enhances Th1 type immune response and anti-tumor immunity against R. Anandan, M. Gopalakrishnan, S. Nagini, Investigation of chemopreventive
breast tumor associated antigen, Cancer Immun. 7 (2007) 8. potential of neem leaf subfractions in the hamster buccal pouch model and
[35] F.O. Othman, G. Motalleb, S.L.T. Peng, A. Rahmat, R. Basri, C.P. Pei, Effect of phytochemical characterization, Eur. J. Med. Chem. 56 (2012) 271–281.
neem leaf extract (Azadirachta indica) on c-myc oncogene expression in 4T1 [57] K.K. Kishore, R. Ganugula, D.R. Gade, G.B. Reddy, S. Nagini, Gedunin
breast cancer cells of Balb/c mice, Cell J. 14 (2012) 53–60. abrogates aldose reductase, PI3 K/Akt/mToR, and NF-kB signaling pathways
[36] P. Kundu, S. Barik, K. Sarkar, A. Bose, R. Baral, S. Laskar, Chemical to inhibit angiogenesis in a hamster model of oral carcinogenesis, Tumor
investigation of neem leaf glycoprotein used as immunoprophylatic agent Biol (2015) (Sep 5. [Epub ahead of print] PMID: 26342697).
for tumor growth restriction, Int. J. Pharm. Pharm. Sci. 7 (2015) 195–199. [58] R. Subapriya, S. Nagini, Ethanolic neem leaf extract protects against
[37] S. Banerjee, T. Ghosh, S. Barik, A. Das, S. Ghosh, A. Bhuniya, A. Bose, R. Baral, N-methyl-N’nitro-N-nitroguanidine-induced gastric carcinogenesis in
Neem leaf glycoprotein prophylaxis transduces immune dependent stop Wistar rats, Asian Pac. J. Cancer Prev. 4 (2003) 215–223.
signal for tumor angiogenic switch within tumor microenvironment, PLoS [59] S. Arivazhagan, B. Velmurugan, V. Bhuvaneswari, S. Nagini, Effects of
One 9 (2014) e110040. aqueous extracts of garlic (Allium sativum) and neem (Azadirachta indica)
[38] A. Bose, E. Haque, R. Baral, Neem leaf preparation induces apoptosis of leaf on hepatic blood oxidant-antioxidant status during experimental
tumor cells by releasing cytotoxic cytokines from human peripheral blood gastric carcinogenesis, J. Med. Food 7 (2004) 334–339.
mononuclear cells, Phytother. Res. 21 (2007) 914–920. [60] T. Dasgupta, S. Banerjee, P.K. Yadava, A.R. Rao, Chemopreventive potential of
[39] S. Goswami, A. Bose, K. Sarkar, S. Roy, T. Chakraborty, U. Sanyal, R. Baral, Azadirachta indica (neem) leaf extract in murine carcinogenesis model
Neem leaf glycoprotein matures myeloid derived dendritic cells and systems, J. Ethnopharmacol. 92 (2004) 23–36.
optimizes anti-tumor T cell functions, Vaccine 28 (2010) 1241–1252. [61] S.C. Gangar, R. Sandhir, D.V. Rai, A. Koul, Modulatory effects of Azadirachta
[40] T. Chakraborty, A. Bose, K.K. Goswami, S. Goswami, K. Chakraborty, R. Baral, indica on benzo(a)pyrene-induced forestomach tumorigenesis in mice,
Neem leaf glycoprotein suppresses regulatory T cell mediated suppression World J. Gastroenterol. 12 (2006) 2749–2755.
of monocyte/macrophage functions, Int. Immunopharmacol. 12 (2012) [62] M.E.T. Manal, P. Hanachi, I. Patimah, I.A. Siddig, O. Fauziah, The effect of
326–333. neem (Azadirachta indica) leaves extract on alpha-fetoprotein serum
[41] K.K. Goswami, S. Barik, M. Sarkar, A. Bhowmick, J. Biswas, A. Bose, R. Baral, concentration, glutathione S-transferase and glutathione peroxidase activity
Targeting STAT3 phosphorylation by neem leaf glycoprotein prevents in hepatocarcinogenesis induced rats, Int. J. Cancer Res. 3 (2007) 111–118.
immune evasion exerted by supraglottic laryngeal tumor induced M2 [63] M.M.E. Taha, S.I.A. Wahab, F. Othman, P. Hanachi, A. Bustamam, A.S.
macrophages, Mol. Immunol. 2 (2014) 119–127. Al-Zubairi, In vivo anti-tumor effects of Azadirachta indica in rat liver cancer,
[42] S. Jafari, S. Saeidnia, H. Hajmehdipoor, M.R.S. Ardekani, M.A. Faramarzi, A. Res. J. Biol. Sci. 4 (2009) 48–53.
Hadjiakhoondi, M. Khanavi, Cytotoxic evaluation of Melia azedarach in [64] A. Ramzanighara, F. Ezzatighadi, D.V. Rai, D.K. Dhawan, Effect of neem
comparison with, Azadirachta indica and its phytochemical investigation, (Azadirichta indica) on serum glycoprotein contents of rats administered
DARU J. Pharm. Sci. 21 (2013) 37. 1,2-dimethylhydrazine, Toxicol. Mech. Methods 19 (2009) 298–301.
[43] A. Roma, P. Ovadje, M. Steckle, L. Nicoletti, A. Saleem, J.T. Arnason, S. Pandey, [65] S.C. Gupta, S. Prasad, D.R. Sethumadhavan, M.S. Nair, Y. Mo, B.B. Aggarwal,
Selective induction of apoptosis by Azadirachta indica leaf extract by Nimbolide a limonoid triterpene, inhibits growth of human colorectal
targeting oxidative vulnerabilities in human cancer cells, J. Pharm. Sci. 4 cancer xenografts by suppressing the proinflammatory microenvironment,
(2015) 729–746. Clin. Cancer Res. 19 (2013) 4465–4476.
[44] K. Kavitha, R. Vidya Priyadarsini, P. Anitha, K. Ramalingam, R. Sakthivel, G. [66] A. Das, S. Barik, A. Bose, S. Roy, J. Biswas, R. Baral, S. Pal, Murine carcinoma
Purushothaman, A.K. Singh, D. Karunagaran, S. Nagini, Nimbolide, a neem expressing carcinoembryonic antigen-like protein is restricted by antibody
liminoid abrogates canonical NF-kB and Wnt signaling to induce against neem leaf glycoprotein, Immunol. Lett. 162 (2014) 132–139.
caspase-dependent apoptosis in human hepatocarcinoma (HepG2) cells, [67] G.H. Kumar, K.V.P.C. Mohan, A.J. Rao, S. Nagini, Nimbolide a limonoid from
Eur. J. Pharmacol. 681 (2012) 6–14. Azadirachta indica inhibits proliferation and induces apoptosis of human
[45] S.C. Gupta, S. Reuter, K. Phromnoi, B. Park, P.S. Hema, M. Nair, B.B. Aggarwal, choriocarcinoma (BeWo) cells, Invest. New Drugs 27 (2009) 246–252.
Nimbolide sensitizes human colon cancer cells to TRAIL through reactive [68] R.V. Priyadarsini, R.S. Murugan, P. Sripriya, D. Karunagaran, S. Nagini, The
oxygen-specie- and ERK-dependent up-regulation of death receptors, p53, neem limonoids azadirachtin and nimbolide induce cell cycle arrest and
and Bax, J. Biol. Chem. 2 (2011) 1134–1146. mitochondria-mediated apoptosis in human cervical cancer (HeLa) cells,
[46] S. Babykutty, P.S. Priya, R.J. Nandini, M.A.S. Kumar, M. Nair, P. Srinivas, S. Free Radic. Res. 44 (2010) 624–634.
Gopala, Nimbolide retards tumor cell migration, invasion, and angiogenesis [69] S. Roy, S. Barik, S. Banerjee, A. Bhuniya, S. Pal, P. Basu, J. Biswas, S. Goswami,
by downregulating MMP-2/9 expression via inhibiting ERK1/2 and reducing T. Chakraborty, A. Bose, R. Baral, Neem leaf glycoprotein overcomes
DNA-binding activity of NF-kB in colon cancer cells, Mol. Carcinog. 51 indoleamine 2,3-dioxygenase mediated tolerance in dendritic cells by
(2012) 475–490. attenuating hyperactive regulatory T cells in cervical cancer stage IIIB
[47] P. Srivastava, N. Yadav, R. Lella, A. Schneider, A. Jones, T. Marlowe, G. Lovett, patients, Hum. Immunol. 74 (2013) 1015–1023.
K. O’Loughlin, H. Minderman, R. Gogada, D. Chandra, Neem oil liminoids [70] M.K. Roy, M. Kobori, M. Takenaka, K. Nakahara, H. Shinmoto, S. Isobe, T.
induces p53-independent apoptosis and autophagy, Carcinogenesis 33 Tsushida, Antiproliferative effect of human cancer cell lines after treatment
(2012) 2199–2207. with nimbolide extracted from an edible part of the neem tree, Phytother.
[48] N. Yadav, S. Kumar, R. Kumar, P. Srivastava, L. Sun, P. Rapali, T. Marlowe, A. Res. 21 (2007) 245–250.
Schneider, J.R. Inigo, J. O’Malley, R. Londonkar, R. Gogada, A.K. Chaudhary, D. [71] S.C. Gupta, S. Prasad, S. Reuter, R. Kannappan, V.R. Yadav, J. Ravindran, P.S.
Yadava Chandra, Mechanism of neem liminoids-induced cell death in Hema, M.M. Chaturvedi, M. Nair, B.B. Aggarwal, Modification of cysteine 179
cancer: role of oxidative phosphorylation, Free Radic. Biol. Med. 90 (2016) of IkBa kinase by nimbolide leads to down-regulation of NF-kB-regulated
261–271. cell survival and proliferative proteins and sensitization of tumor cells to
[49] B.S. Sastry, S. Babu, T.H. Babu, S. Chandrasekar, P.V. Srinivas, A.K. Saxena, M. chemotherapeutic agents, J. Biol. Chem. 46 (2010) 35406–35417.
Rao, Synthesis and biological activity of amide derivatives of nimbolide, [72] T. Kikuchi, K. Ishii, T. Noto, A. Takahashi, K. Tabata, T. Suzuki, T. Akhisa,
Bioorg. Med. Chem. Lett. 16 (2006) 4391–4394. Cytotoxic apoptosis-inducing activities of liminoids from the seeds of
[50] S. Balasenthil, S. Arivazhagan, C.R. Ramachandran, V. Ramachandran, S. Azadirachta Indica (neem), J. Nat. Prod. 74 (2011) 866–870.
Nagini, Chemopreventive potential of neem (Azadirachta indica) on [73] S. Kumar, P.K. Suresh, M.R. Vijayababu, A. Arunkumar, J. Arunakaran,
7,12-dimethylbenz[a]anthracene (DMBA) induced hamster buccal pouch Anticancer effects of ethanolic neem leaf extract on prostate cancer cell line
carcinogenesis, J. Ethnopharmacol. 67 (1999) 189–195. (PC-3), J. Ethnopharmacol. 105 (2006) 246–250.
[51] R. Subapriya, V. Bhuvaneswari, S. Nagini, Ethanolic neem (Azadirachta [74] D.N. Gunadharini, P. Elumalai, R. Arunkumar, K. Senthilkumar, Arunakaran
indica) leaf extract induces apoptosis in the hamster buccal pouch Induction of apoptosis and inhibition of PI3K-Akt pathway in PC-3 and
carcinogenesis model of modulation of Bcl-2, Bim, caspase 8 and caspase 3, LNCaP prostate cancer cells by ethanolic neem leaf extract, J.
Asian Pac. J. Cancer Prev. 6 (2005) 515–520. Ethnopharmacol. 134 (2011) 644–650.
[52] R. Subapriya, R. Kumaraguruparan, S. Nagini, Expression of PCNA, [75] S. Mahapatra, R.J. Karnes, M.W. Holmes, C.Y.F. Young, J.C. Cheville, M. Kohli,
cytokeratin, Bcl-2 and p53 during chemoprevention of hamster buccal E.W. Klee, D.J. Tindall, K.V. Donkena, Novel molecular targets of Azadirachta
pouch carcinogenesis by ethanolic neem (Azadirachta indica) leaf extract, indica associated with inhibition of tumor growth in prostate cancer, APPS J.
Clin. Biochem. 39 (2006) 1080–1087. 13 (2011) 365–377.
[53] P. Manikandan, P.V. Letchoumy, M. Gopalakrishnan, S. Nagini, Evaluation of [76] Q. Wu, M. Kohil, H.R. Bergen III, J.C. Cheville, R.J. Karnes, H. Cao, C.Y.F. Young,
Azadirachta indica leaf fraction for in vitro antioxidant potential and in vivo D.J. Tindall, M.A. McNiven, K.V. Donkena, Preclinical evaluation of the
modulation of biomarkers of chemoprevention in the hamster buccal pouch supercritical extract of Azadirachta Indica (neem) leaves in vitro and in vivo
carcinogenesis model, Food Chem. Toxicol. 46 (2008) 2332–2343. inhibition of prostate cancer tumor growth, Mol. Cancer Ther. 13 (2014)
[54] R.V. Priyadarsini, P. Manikandan, G.H. Kumar, S. Nagini, The neem limonoids 1066–1077.
azadirachta and nimbolide inhibit hamster cheek pouch carcinogenesis by [77] P.R. Singh, R. Arunkumar, V. Sivakamasundari, G. Sharmila, P. Elumalai, E.
modulating xenobiotic-metabolizing enzymes, DNA damage, antioxidants, Suganthanpriya, A.B. Mercy, K. Senthilkumar, J. Arunakumar,
invasion and angiogenesis, Free Radic. Res. 43 (2009) 492–504. Anti-proliferative and apoptosis inducing effect of nimbolide by altering
[55] G.H. Kumar, V. Priyadarsini, G. Vinothini, V. Letchoumy, S. Nagini, The neem molecules involved in apoptosis and IGF signaling via PI3K/Akt in prostate
liminoids azadirachtin and nimbolide inhibit cell proliferation and induce cancer (PC-3) cell line, Cell Biochem. Funct. 32 (2014) 217–228.
apoptosis in an animal model of oral oncogenesis, Invest. New Drugs 28 [78] J. Zhang, K.S. Ahn, C. Kim, M.K. Shanmugam, K.S. Siveen, F. Arfuso, R.P. Samy,
(2010) 392–401. A. Deivasigamani, L.H. Lim, L. Wang, B.C. Goh, A.P. Kumar, K.M. Hui, G. Sethi,
Nimbolide-induced oxidative stress abrogates STAT3 signaling cascade and

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002
G Model
YSCBI-1243; No. of Pages 16 ARTICLE IN PRESS
16 S.M. Patel et al. / Seminars in Cancer Biology xxx (2016) xxx–xxx

inhibits tumor growth in transgenic adenocarcinoma of mouse prostate perforatum and neem oil for the management of acute skin toxicity in head
model, Antioxid. Redox Signal. (2015) (Dec 9. [Epub ahead of print] PMID: and neck cancer patients undergoing radiation or chemo-radiation: a
26649526). single-arm prospective observational study, Radiat. Oncol. 9 (2014) 297.
[79] T. Akihisa, T. Noto, A. Takahashi, Y. Fujita, N. Banno, H. Tokuda, K. Koike, T. [89] S.J. Boeke, M.G. Boersma, G.M. Alink, J.J.A. Van Loon, A. Van Huis, M. Dicke,
Suzuki, K. Yasukawa, Y. Kimura, Melanogenesis inhibitory, Rietjens IMCM. Safety evaluation of neem (Azadirachta indica) derived
anti-inflammatory, and chemopreventive effects of liminoids from the seeds pesticides, J. Ethnopharmacol. 94 (2004) 25–41.
of Azadirachta indica A. Juss. (Neem), J. Oleo Sci. 58 (2009) 581–594. [90] S.N. Okpanyi, G.C. Ezeukwu, Anti-inflammatory and antipyretic activities of
[80] T. Akihisa, A. Takahashi, T. Kikuchi, M. Takagi, K. Watanabe, M. Fukatsu, Y. Azadirachta indica, Planta Med. 41 (1981) 34–39.
Fujita, N. Banno, H. Tokuda, K. Yasukawa, The melanogenesis-inhibitory, [91] P. Kupradinun, A. Tepsuwan, N. Tanthasri, N. Meesiripan, S. Tunsakul, W.
anti-inflammatory, and chemopreventive effects of liminoids in n-hexane Tompat, Y. Jarratwisarutporn, W.R. Kusamran, Toxicity testing of flowers of
extract of Azadirachta indica A. Juss (neem) seeds, J. Oleo Sci. 60 (2011) neem tree, Thai J. Vet. Med. 40 (2010) 47–55.
53–59. [92] M. Dorababu, M.C. Joshi, G. Bhawani, M.M. Kumar, A. Chaturvedi, R.K. Goel,
[81] S. Barik, A. Bhuniya, S. Banerjee, A. Das, M. Sarkar, T. Paul, T. Ghosh, S. Ghosh, Effect of aqueous extract of neem (Azadirachta indica) leaves on offensive
S. Roy, S. Pal, A. Bose, R. Barat, Neem leaf glycoprotein is superior than and diffensive gastric mucosal factors in rats, Indian J. Physiol. Pharmacol.
cisplatin and sunitinib malate in restricting melanoma growth by 50 (2006) 241–249.
normalization of tumor microenvironment, Int. Immunopharmacol. 17 [93] R.B. Raizada, M.K. Srivastava, R.A. Kaushal, R.P. Singh, Azadirachtin a neem
(2013) 42–49. biopesticide: subchronic toxicity assessment in rats, Food Chem. Toxicol. 39
[82] N. Arora, A. Koul, M.P. Bansal, Chemopreventive activity of Azadirachta (2001) 477–483.
indica on two-stage skin carcinogenesis in murine model, Phytother. Res. 25 [94] F.M.A. Tarboush, H.M.E. Ashamoui, Toxic and teratogenic effects of
(2011) 408–416. Azadirachtin of Neemix-4.5 on fetuses and pups of SWR/J mice, Eyg. J. Hosp.
[83] N. Arora, M.P. Bansal, A. Koul, Azadirachta indica acts as a pro-oxidant and Med. 15 (2004) 30–39.
modulates cell cycle associated proteins during DMBA/TPA induced skin [95] M.K. Srivastava, R.B. Raizada, Lack of toxic effects of technical azadirachtin
carcinogenesis in mice, Cell Biochem. Funct. 31 (2013) 385–394. during postnatal development of rats, Food Chem. Toxicol. 45 (2007)
[84] A. Mallick, S. Barik, K.K. Goswami, S. Banerjee, S. Ghosh, K. Sarkar, A. Bose, R. 465–471.
Baral, Neem leaf glycoprotein activates CD8+ T cells to promote therapeutic [96] Farm Chemicals Handbook, Meister Publishing Co., Willoughby, OH, (1995).
anti-tumor immunity inhibiting the growth of mouse sarcoma, PLoS One 8 [97] T. Glinsukon, P. Somjaree, P. Piyachaturawat, Y. Thebtaranonth, Acute
(2013) e47434. toxicity of nimbolide and nimbic acid in mice, rats and hamsters, Toxicol.
[85] S. Barik, S. Banerjee, A. Mallick, K.K. Goswami, S. Roy, A. Bose, R. Baral, Lett. 30 (1986) 159–166.
Normalization of tumor microenvironment by neem leaf glycoprotein [98] M.R. Gandhi, R. Lal, A. Sankaranarayanan, C.K. Banerjee, P.L. Sharma, Acute
potentiates effector T cell functions and therapeutically intervenes in the toxicity study of the oil from Azadirachta indica seed (neem oil), J.
growth of mouse sarcoma, PLoS One 8 (2013) e66501. Ethnopharmacol. 23 (1988) 39–51.
[86] A. Mallick, S. Barik, S. Ghosh, S. Roy, K. Sarkar, A. Bose, R. Baral, [99] P.R. Karmakar, B.P. Chatterjee, Isolation and characterization of two
Immunotherapeutic targeting of established sarcoma in Swiss mice by IgE-reactive proteins from Azadirachta indica pollen, Mol. Cell. Biochem. 131
tumor-derived antigen-pulsed NLGP matured dendritic cells is CD8+ T-cell (1994) 87–96.
dependent, Immunotherapy 7 (2014) 821–831. [100] J. Ferlay, I. Soerjomataram, M. Ervik, R. Dikshit, S. Eser, C. Mathers, et al.,
[87] A. Vasenwala, R. Seth, N. Haider, N. Islam, T. Khan, V. Maheshwari, S. GLOBOCAN v1.0 (2012), Cancer Incidence and Mortality Worldwide, IARC
Rehman, A study on antioxidant and apoptotic effect of Azadirachta Indica CancerBase No.11. Lyon, France, International Agency for Research on
(neem) in cases of cervical cancer, Arch. Gynecol. Obstet. 286 (2012) Cancer, Available from http://globocan.iarc.fr, 2013 (accessed 22.2.16)
1255–1259. [Internet].
[88] P. Franco, I. Potenza, F. Moretto, M. Segantin, M. Grosso, A. Lombardo, D.
Taricco, P. Vallario, A.R. Filippi, M. Rampino, U. Ricardi, Hypericum

Please cite this article in press as: S.M. Patel, et al., Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic
diseases, Semin Cancer Biol (2016), http://dx.doi.org/10.1016/j.semcancer.2016.03.002

You might also like