ArticlePDF AvailableLiterature Review

Nanoformulations of Coumarins and the Hybrid Molecules of Coumarins with Anticancer Effects

Authors:

Abstract and Figures

Coumarins are the seconder metabolites of some plants, fungi, and bacteria. Coumarins and the hybrid molecules of coumarins are the compounds which have been widely studied for their anticancer effects. They belong to benzopyrone chemical class, more precisely benzo-α-pyrones, where benzene ring is fused to pyrone ring. In nature, coumarins are found in higher plants like Rutaceae and Umbelliferae and some essential oils like cinnamon bark oil, cassia leaf oil and lavender oil are also rich in coumarins. The six main classes of coumarins are furanocoumarins, dihydrofuranocoumarins, pyrano coumarins, pyrone substituted coumarins, phenylcoumarins and bicoumarins. As well as their wide range of biological activities, coumarins and the hybrid molecules of coumarins are proven to have an important role in anticancer drug development due to the fact that many of its derivatives have shown an anticancer activity on various cell lines. Osthol, imperatorin, esculetin, scopoletin, umbelliprenin, angelicine, bergamottin, limettin, metoxhalen, aurapten and isopimpinellin are some of these coumarins. This review summarizes the anticancer effects of coumarins and their hybrid molecules including the novel pharmaceutical formulations adding further information on the topic for the last ten years and basically focusing on the structure-activity relationship of these compounds in cancer.
Content may be subject to copyright.
Anti-Cancer Agents in Medicinal Chemistry
The journal for in-depth reviews and high quality research
papers on Anti-Cancer Agents
Impact
Factor:
2.04
ISSN: 1871-5206
eISSN: 1875-5992
Send Orders for Reprints to reprints@benthamscience.net
Anti-Cancer Agents in Medicinal Chemistry, 2020, 20, 1797-1816
1797
REVIEW ARTICLE
Nanoformulations of Coumarins and the Hybrid Molecules of Coumarins with
Potential Anticancer Effects
Mukerrem Betul Yerer1,2,*, Serkan Dayan2, M. Ihsan Han3, Ajay Sharma4, Hardeep S. Tuli5 and Katrin Sak6
1Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri, Turkiye; 2Drug Application and Research Center,
Erciyes University, Kayseri, Turkey; 3Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri,
Turkiye; 4Department of Chemistry, Career Point University, Tikker-kharwarian, Hamirpur, Himachal Pradesh 176041, India;
5Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana-133207, India;
6NGO Praeventio, Tartu, Estonia
A R T I C L E H I S T O R Y
Received: October 07 , 2019
Revised: November 04, 2019
Accepted: November 28, 201 9
DOI:
10.2174/187152062066620031009464
6
Abstract: Coumarins are the secondary metabolites of some plants, fungi, and bacteria. Coumarins and the hybrid
molecules of coumarins are the compounds which have been widely studied for their potential anticancer effects.
They belong to benzopyrone chemical class, more precisely benzo-α-pyrones, where benzene ring is fused to pyrone
ring. In nature, coumarins are found in higher plants like Rutaceae and Umbelliferae and some essential oils like
cinnamon bark oil, cassia leaf oil and lavender oil are also rich in coumarins. The six main classes of coumarins are
furanocoumarins, dihydrofuranocoumarins, pyrano coumarins, pyrone substituted coumarins, phenylcoumarins and
bicoumarins. As well as their wide range of biological activities, coumarins and the hybrid molecules of coumarins
are proven to have an important role in anticancer drug development due to the fact that many of its derivatives have
shown an anticancer activity on various cell lines. Osthol, imperatorin, esculetin, scopoletin, umbelliprenin, an-
gelicine, bergamottin, limettin, metoxhalen, aurapten and isopimpinellin are some of these coumarins. This review
summarizes the anticancer effects of coumarins and their hybrid molecules including the novel pharmaceutical for-
mulations adding further information on the topic for the last ten years and basically focusing on the structure-
activity relationship of these compounds in cancer.
Keywords: Coumarines, coumarine derivatives, coumarine nanoformulations, coumarine hybride molecules, anticancer effect, metabolites.
1. INTRODUCTION
The incidence of malignant disorders is increasing at an alarm-
ing rate worldwide. Although several therapeutically active molecules
have been investigated in the last decade, the curative treatment is
still not defined for many cancer types, especially for advanced and
metastatic malignancies. Moreover, the existing anti-cancer strate-
gies are known to possess a variety of side effects, while cancerous
cells may be or become resistant towards these therapies.
The existing anti-cancer strategies are known to possess a vari-
ety of side effects [1, 2]. Therefore, one of the greatest challenges
for the scientific community is to design novel therapeutic agents to
fight against cancer. Evidence has suggested that various plant
originated secondary metabolites such as coumarins, alkaloids,
terpenoids, phenolic, and polyphenols compounds possess promis-
ing anti-cancer activity [3, 4].
Coumarins consist of seven main classes including furanocou-
marins, dihydrofuranocoumarins, pyrano coumarins, pyrone substi-
tuted coumarins, phenylcoumarins and bicoumarins. Structurally
coumarins are classified under benzopyrone family with fused ben-
zene and pyrone ring [5, 6]. Furthermore, investigations are in pro-
gress to synthesize novel coumarins hybrid molecules with im-
proved anticancer activity and bioavailability [7-12].
However, due to the low bioavailability, the role of biologically
active moieties in the treatment of cancer is still undermined [13].
Therefore, to enhance the bio-availability and potency of drug,
*Address correspondence to this author at the Department of Pharmacology,
Faculty of Pharmacy, Drug Application and Research Center, Erciyes Uni-
versity, P.O. Box: 38039, Kayseri, Turkey; Tel/Fax: +903522076666/13852;
E-mail: mbyerer@erciyes.edu.tr
nano-formulation system could be a promising tactic. Recent inves-
tigations have stated that nano-formulations based on polymeric
micelles, polymeric brush, dendrimers, nanogels, and liposomes are
studied widely for drug delivery in the biological systems [14, 15].
Coumarins are found to upregulate and downregulate apoptotic
(Bax, caspase-3) and anti-apoptotic (Bcl-2) proteins, respectively
[16, 17]. Cell cycle regulatory proteins are also downregulated by
coumarins, which lead to the G0/G1 cell cycle arrest in cancer [17].
In addition, coumarins treatment results in the inhibition of angio-
genesis and metastasis in cancer via the downregulation of VEGF
and MMPs expression [18, 19].
Studies have revealed that coumarins suppress the expression of
inflammatory mediators such as iNOS and COX-2 by inhibiting
NF-κB, MAPK and Akt signaling [20]. It has been seen that phyto-
chemicals present in nanoformulation possess better biological
activity as compared to their native forms [13, 14]. Therefore, the
present review d escribes recent advancement in the nanoformula-
tions of coumarins and their derivatives against cancer.
1.1. Structural Features of Coumarins and Coumarin
Derivatives
Coumarins consist of six main classes including furanocoumar-
ins, dihydrofuranocoumarins, pyrano coumarins, pyrone substituted
coumarins, phenylcoumarins and bicoumarins (Table 1). They can
be either classified as:
Benzene-Substituted Coumarins
Pyrone-Substituted Coumarins
Both benzene-substituted and pyrone-substituted coumarins
Coumarins obtained by bonding other ring structures to the
benzene ring or the pyron ring,
1875-5992/20 $65.00+.00 © 2020 Bentham Science Publishers
1798 Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 Yerer et al.
Bicoumarins
Furanocoumarins
Each coumarin sub-type is studied and valued by researchers.
As is known, coumarin derivatives have many application areas due
to their biological and chemical properties [21], which can be listed
as anticancer [22-27], carbonic anhydrase inhibitors [28-30], anti-
HIV [31-33], anti-inflammatory [34-36] fluorescence [37, 38] and
catalysis chemistry [39, 40].
These structures, which have many different features, both con-
tribute to technological developments and take place in many sys-
tems as qualified chemicals. In this context, coumarins, which are
also usable in nano-technological developments, have also contrib-
uted to nanoformulation or nanocapsulation studies in recent years
(Fig. 1).
Nanostructures and the guest molecules are the basis of nano-
formulation studies. The basis of nanoformulation studies is divided
into two main categories; guest molecules and nanocarriers. The
nanocarriers with some nanostructures such as multifunctional
nanocapsules, have the character of containers that can be loaded
with various cargoes and then become promising nanocarriers for
diagnostic and drug delivery, or some biomedical applications.
Such Nanoparticles (NPs) or nanocarriers can be charged with
many guest molecules, such as hydrophobic therapeutics, optically
active organic molecules, metal complexes, ultrasonic contrast
agents, metal oxides, etc. and the particles may also change the
potential solubility problems and biocompatibility of the guest
molecules.
The present positions of the manufactured nanoparticles allow
for combining different properties for the drug delivery and diag-
nostic systems. Thus, the guest molecules can be transported to the
desired sites and however, some modifications to the particles may
be required such as shape, stability, physical state, charge, perme-
ability, environmental responsiveness etc. In addition to the combi-
nation of all these properties, the nanocarrier and the guest mole-
cules must be in harmony and serve the same target. The coumarins
are also u sed in many research guest molecules and the use of cou-
marins in nanomaterials is also increasing (Fig. 2).
In the drug delivery system, there are mainly carrier particles,
guest molecules and support molecules for detecting/imaging the
system. In addition to being guest molecules, the coumarin and
their derivative compounds can also serve to visualize the formula-
tions. In this sense, the Coumarin-6 is one of the most commonly
used compounds in formulation studies [41-46]. This compound is
one of both benzene-substituted and pyrano-substituted coumarin
derivatives and has been used as a fluorescence molecule for mark-
ing NPs in many formulation studies.
By Paiva et al., a novel nanoformulation for p53-MDM2 inter-
action inhibitor was formed using several techniques and the toxic
potentials of the materials were investigated on MCF-7 cells. In this
Table 1. Some main coumarin structures.
Simple Coumarins
O O
R
Furanocoumarins
O O
O
Psoralen
O O
O
Xanthotoxin
OCH
3
Pyranocoumar ins
OO
Xa nthylet in
O O
Sesel in
O
Benzene-Substituted Co umarins
O O
H
3
CO
HO
Scopoletin
O O
H
3
CO
HO
Fraxetin
OH
Pyrone-Substituted Coumarins
O O
OH
O
Warfarin
O O
OH
Phenprocoumon
Bicoumarins
O
O
O
O
OHHO
C
H
2
Dicoumarol
Nanoformulations of Coumarins and the Hybrid Molecules of Coumarins Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 1799
study, the monitoring of nanoparticle penetration into the cell was
carried out by monitoring the coumarin-6 green fluorescence mole-
cule [47].
Bazylinska et al. reported the encapsulation of chemotherapeu-
tic (colchicine) and imaging agent (coumarin-6) in oil-core nanocar-
riers stabilized by surfactants and the cytotoxicity potentials were
evaluated by CLSM analysis on some human cancer cells such as
skin melanoma (MEWO), doxorubicin-sensitive breast cancer
(MCF-7/WT), and alveolar basal epithelial cells (A549) [48].
Zhao and co-workers fabricated Paclitaxel-loaded (PTX-
loaded) [PGA-co-PCL]-b-TPGS2k nanoformulation for lung cancer
therapy and the antitumor activity of the nanomaterial was exam-
ined both in vitro and in vivo on human lung cancer A549 cells.
Herein, the coumarin-6 in place of PTX was encapsulated and in
this way, Confocal Laser Scanning Microscopy (CLSM) images of
tumor cells were observed [49].
Su and co-workers recorded the Paclitaxel (PTX)-loaded star-
shaped copolymer bearing coumarin-6 as fluorescence biomarker
Fig. (1). Evolution of number of publications for ‘nanoformulation or nanocapsulation’ keywords. Source: ISI Web of Science, Thomson Reuters. (A higher
resolution / colour version of this figure is available in the electronic copy of the article).
Fig. (2). Evolution of publication number for ‘coumarin and nano’ keywords. Source: ISI Web of Science, Thomson Reuters. (A higher resolution / colour
version of this figure is available in the electronic copy of the article).










ϭϬϬϬϬϬϮϬϭϱϱ
ϭϮ
Ϯϯ
ϰϲ ϰϳ
ϴϰ
ϭϭϮ ϭϭϴ
ϭϲϴ
ϭϴϵ

   





ϭϮ
Ϭϭϭ
ϯ
ϭ
ϰϰ
ϲϱ
ϭ
ϭϮ
ϵ
Ϯϯ
ϭϰ ϭϱ
ϮϬ
Ϯϲ
ϮϮ

   
1800 Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 Yerer et al.
nanoformulation for improving chemotherapy of Malignant Mela-
noma (MM), which is the dangerous skin cancer type. The anti-
tumor efficiency of the fabricated nanomaterial, studied in vitro
with the MTT assay and in vivo with xenograft tumor model,
showed that the star-shaped PTX-loaded CA-PLGA-TPGS nano-
material was importantly superior to commercial PTX formulation
[50].
Kushwah et al. recorded the self-assembled nanomaterial for
improved breast cancer therapy with the dual drug conjugation
which included Docetaxel (DTX) and Gemcitabine (GEM). Simi-
larly, the coumarin-6 (3-2′-benzothiazolyl-7-diethylaminocoumarin)
was used as a model fluorescent dye. Herein, the results demon-
strated significant decrease in both the volume of tumor and hepato-
and nephro-toxicity [51].
Although the coumarin-6 fluorescence molecule is used
frequently, other coumarin molecules such as coumarin-153,
coumarin-314 or coumarin-blue, which have similar properties and
are used in nanoformulation studies, are also used as fluorescence
biomarkers (Table 2). These molecules, like coumarin-6, are both
benzene substituted and pyrano substituted coumarin derivatives
[52-54].
In addition to these studies, some synthetic benzene-substituted
coumarin-derived compounds have found their place in nanoformu-
lation studies as guest molecules. Lee and co-workers demonstrated
that self-assembled coumarin (both benzene-substituted and py-
rone-substituted coumarins) nanoparticles have anti-cancer poten-
tials with selective mitochondrial-targeting. Herein, the encapsula-
tion of cancer therapy drug Doxorubicin (DOX) into the fabricated
Triphenylphosphonium-appended Coumarin nanomaterial (TPP-C),
which showed non-cytotoxicity toward cancer cell line, was
achieved and determined that the doxorubicin was effectively de-
livered to the mitochondria. This study also clearly demonstrated
that such a drug delivery system is the preferred system for the
treatment of cancer [55].
1.2. Classification of Coumarin Derivatives and Hybrid
Molecules under Several Cancer Types
1.2.1. Lung Cancer
Maiti et al. designed, synthesized and characterized gemcit-
abine-coumarin-biotin conjugate for in vitro biological evaluation.
This molecule is a multifunctional molecule composed of a thiol-
specific cleavable disulfide bond, a coumarin moiety as a fluores-
cent reporter, Gemcitabine (GMC) as a model active drug, and
biotin as a cancer-targeting unit. The active molecule is preferen-
tially taken up by A549 cells rather than WI38 cells according to
the Confocal microscopic experiments. This drug delivery system is
a novel theranostic agent. By two photon fluorescence imaging,
both a therapeutic effect and drug uptake could b e monitored [56].
In their study, Khaghanzadeh et al. researched the impact of
umbelliprenin on the predominance of Th1 and Th2 responses in
the Lewis Lung Cancer (LLC) mouse model. The cytotoxic effect
of umbelliprenin (7-prenyloxycoumarins) was discovered on LLC
cells and mouse splenocytes by MTT assay. On normal splenocytes,
results displayed the cytotoxic effects of umbelliprenin on LLC cells
(IC50= 51.6±5.4µM), whereas no side effects were screened at this
concentration. TNF-α mRNA expression in both lung and tumor
tissues was increased. Umbelliprenin has displayed a partly Th1
preconception with a reduction in regulatory immune response [57].
Table 2. Some fluorescence coumarins used in nano formulation st udies.
Chemical Name Chemical Structure
3-(2-Benzothiazolyl)-7-(diethylamino)
coumarin
O O
S
N
N
CH
3
CH
3
Coumarin-6
2,3,6,7-Tetrahydro-9-trifluoromethyl-1H,5H-quinolizino
(9,1-gh)coumarin
O O
Coumarin-153
CF
3
N
2,3,5,6-1H,4H-Tetrahydr o-9-carbetho xyquinolizino-
[9,9a,1-gh]coumarin
O ON
O
OCH
3
Coumarin-314
7-(diethylamino)-coumarin-3-carboxylic
acid-N-succinimidyl ester
O ON
CH
3
CH
3
Coumarin-blue
O
O
N
O
O
Nanoformulations of Coumarins and the Hybrid Molecules of Co umarins Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 1801
1.2.2. Breast Cancer
Because of their skill to ensure spatial and temporal control
over the drug release, photoresponsive nanoparticles have admitted
important care. Photoresponsive multifunctional Magnetic
Nanoparticles (MNPs) were noticed utilizing coumarin-based pho-
totrigger and Fe/Si MNPs for checked delivery of anticancer drug
chlorambucil. Produced photoresponsive multifunctional MNPs
were also discovered for cell luminescence imaging. In vitro bio-
logical works displayed that coumarin connected Fe/Si MNPs of
9nm size d elivered the anticancer drug chlorambucil into cancer
cells, thereby influencing the drug action to kill the cancer cells
upon irradiation. Cell viability was evaluated utilizing MTT assay
in breast cancer cell line MDA-MB-231. Cells were incubated with
chlorambucil, chlorambucil-loaded MNPs (7-HC-Cbl-MNPs), and
Fe/Si MNPs for 4h over a range of concentrations between 0 and
110μM. It was considered that IC50 values of MDA-MB-231 cells
reduced from 67.38±1.45μM for chlorambucil to 53.84±1.12μM for
the 7-HC-Cbl-MNPs [58].
Dlugosz et al. appraised anticancer features of a novel synthetic
hybrid compound AD-013, which associates a coumarin moiety and
an α-methylene-δ-lactone subject with a natural antibiotic novobio-
cin. They assessed cytotoxic activities of compound AD-013 using
MTT assay and found the mechanism of anticancer activity of ana-
log AD-013 with RT-PCR. Against two cancer cells, MCF-7 (breast
cancer) and HL-60 (leukemia) and normal human cells MCF-10A
and HUVEC, AD-013 was much more cytotoxic than novobiocin in
both cancer cell lines and showed some selectivity against MCF-7
cancer cells as compared with MCF-10A healthy breast cells [59].
1.2.3. Prostate Cancer
Lalitha et al. synthesized and characterized three distinct cou-
marin-pyrene derivatives with changing hydrophobic connections.
Among these derivatives, the one which is not having any hydro-
phobic tail showed effective gelation. The investigation of absorp-
tion and emission spectra of these compounds displayed that in
dissimilar solvents, the photo-physical properties were significantly
impacted by the self-assembly protocol. They derived nano-flakes
from coumarin coupled pyrene derivatives. They researched their
potential applications in the field of cell imaging by taking a hint
from the self-assembly mechanism in dissimilar solvents. For fibro-
blast and PC3 prostate cancer cell imaging applications, the nano-
materials got utilized via the self-assembly protocol. Investigation
revealed that these molecules decreased the proliferation of PC3
cells [60].
1.2.4. Hepatocellular Carcinoma
Ji et al. designed a fluorescent and multi-functional gelator
precursor with high photosensitivity. Between overexpressed galac-
tose and ASGP-R, cancer cells could be selectively targeted owing
to the receptor mediated interaction by cancer cells. The precursor
could release hydrogelators inside cells under photo-irradiation via
leading to intracellular self-assembly and inducing cell death. With
near infrared light (non-invasive wavelength), coumarin derivatives
had the feature of two-photon absorption, facilitating irradiation of
the precursors which is going on. In order to assess the anticancer
effects, intracellular self-assembly was started by light. CCK-8
assay quantified the viability of both HepG2 and HeLa cells [61].
1.2.5. Ovarian Cancer
By inducing apoptosis and inhibiting cell growth, Docetaxel
(DOC) generates antitumor effects but its clinical practice is limited
by its nominal biocompatibility and hydrophobicity. Through
computer simulations, Gao et al. prepared DOC and methoxy
poly(ethylene glycol) (MPEG)/polycaprolactone (PCL) (MPEG-
PCL) that demonstrated fine compatibility. In vitro DOC/MPEG-
PCL micelles inhibited cell growth. It induced apoptosis more effi-
ciently than free DOC. DOC/MPEG-PCL micelles inhibited ovar-
ian tumor growth more importantly than free DOC. Immunohisto-
chemical analysis indicated that DOC/MPEG-PCL micelles en-
hanced DOC's anti-tumor impact by advancing tumor cell apoptosis
and suppressing tumor cell proliferation [62].
1.2.6. Colon Cancer
Xu et al. designed and synthesized four coumarin-containing
telodendrimers for self-assembling into the corresponding nanopar-
ticles. In order to target drug delivery of 7-ethyl-10-hydroxy camp-
tothecin (SN-38), two nanoparticles (P-II and P-IV micelles) were
viewed and chosen for the treatment of colon cancer. In aqueous
solution, the nanoparticle encapsulation importantly increased the
solubility of SN-38. The size of these SN-38 nanoparticles was
found to be around 50nm and rod-shaped micelles were found util-
izing Transmission Electron Microscopy (TEM) by Dynamic Light
Scattering (DLS). Similar in vitro cytotoxic activities against colon
cancer cells were found for these two novel nanoformulations of
SN-38/P-II and SN-38/P-IV as compared to the basic drug (SN-38
in DMSO). These NPs preferentially accumulated in tumors when
compared with healthy tissue. In HT-29 human colon cancer
xenograft models, SN-38 loaded nanoformulations showed superior
anti-tumor efficacy when compared with Irinotecan at equivalent
SN-38 dose [63].
The possibilities to inhibit a drug's metabolism and thereby
increasing its efficient dose was further evaluated as a new applica-
tion of nanomedicines. Natural furanocoumarin 6′,7′-
Dihydroxybergamottin (DHB), a known CYP450 inhibitor encapsu-
lated in micelles, were explored to transiently block hepatic
CYP450-mediated drug metabolism for the increase in the bioavail-
ability of the oncology drug docetaxel. When compared to the par-
ent drug, DHB-micelles increased antitumor effect in the tumor
xenograft models HT-29 (colon cancer) and MDA-MB-231 (breast
cancer) in mice, when administered 24h prior to the drug [64].
1.2.7. Gastric Cancer
Aas et al. utilized dendrosome nano-particles to overcome the low
bioavailability of farnesiferol C limiting its therapeutic potential.
They evaluated the anticancer impact of Dendrosomal Farnesiferol
C (DFC) on the AGS gastric cancer cell line. In order to define the
antiproliferative features of DFC and expression ratio of Bax/Bcl-2
as a characteristic of apoptosis, MTT assay and Reverse Trans-
criptase-Polymerase Chain Reaction (RT-PCR) were used, respec-
tively. The study demonstrated that DFC importantly suppressed
the proliferation of AGS cells in a time- and dose-dependent man-
ner compared to the void Farnesiferol C (FC). DFC significantly
increased the expression ratio of Bax/Bcl-2 in AGS cells [65].
1.2.8. Melanoma
In their study, Bhattacharya et al. encapsulated 4-methyl-7-
hydroxy coumarin (SC) with Poly Lactide-co-Glycolide Acid
(PLGA) nanoparticles to test if nano-encapsulated SC can be a
more efficient anti-cancer agent. As revealed from scanning elec-
tronic and atomic force microscopies, they studied its potentials
with various protocols. Nanocoumarins (NC) showed more efficient
drug uptake and demonstrated anticancer potentials in melanoma
cell line A375. The combined data of Circular Dichroism spectra
(CD) and melting temperature profile (Tm) of calf thymus DNA
treated with NC were analyzed in order to test its feasible interac-
tion with target DNA. Results indicated a dose dependent interac-
tion of NC with calf thymus DNA, forming a novel complex that
increased its stability and bringing in efficient modification in struc-
ture and conformation [66].
1.2.9. Cervical Cancer
Due to their multiple functionalization features among polym-
eric NPs, star-shaped branched polymers have attracted a great
1802 Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 Yerer et al.
Table 3. Classification of coumarin derivatives and hybrid molecules with anti-cancer effect.
Name and Chemical Structure of Coumarins Pharmacological Activity Refs.
Osthole
7-methoxy-8-(3-methylbut-2-enyl)chromen-2-one
Osthole treatment significantly reduced the expression of the antiapoptotic protein Bcl-2 and
increased the expression of the proapoptotic proteins Bax, Bak, BimL, BimS, and t-Bid. Osthole
treatment also increased the expression of Fas, FADD, TNF-R1, TNF-R2, DcR2, RIP, and DR5.
In addition, osthole treatment significantly increased the expression levels of phosphorylated
ERK1/2 and JNK1/2.
Osthole reduced the levels of matrix metalloproteinase-2 (MMP-2) and matrix metallopepti-
dase-9 (MMP-9) in the A549 human lung cancer cells
Osthole inhibited cells proliferation via inducing apoptosis and G2/M arrest
[72-74]
Imperatorin
9-(3-methylbut-2-enoxy)furo[3,2-g]chromen-7-one
Imperatorin induced a dose-dependent growth inhibition of the SGC-7901 gastric cancer cells
with an IC50 value 62.6μM via cell cycle arrest targeting PI3K/Akt/m-TOR signalling pathway.
Imperatorin inhibited HIF-1α protein synthesis by downregulating the mTOR/p70S6K/4E-BP1
and MAPK pathways in colon cancer.
33 coumarins, mainly the simple isopentenylated coumarins and derived pyrano- and
furanocoumarins, were examined for their antiproliferative activity towards several cancer and
normal human cell lines.
[75-77]
Esculetin
6,7-dihydroxychromen-2-one
Biological studies by dihydroxycoumarins as fluorescent probes performed on prostate cancer
cells, breast cancer cells, and normal skin fibroblasts revealed significant anticancer properties
of esculetin, which caused a considerable decrease in the viability and number of cancer cells,
and affected their morphology, contrary to normal fibroblasts. Furthermore, the experiment per-
formed on prostate cancer cells showed that fluorescence emission of esculetin is closely related
to intracellular pH.
Esculetin exhibits selective anticancer effects in THP-1 human leukemia cells and led to signifi-
cant apoptosis induction, inhibition of cancer cell migration and decrease in Bcl-2/Bax ratio.
Esculetin binds to KEAP1 and inhibits its interaction with Nrf2 in pancreatic cancer cells. This,
thereby, promotes nuclear accumulation of Nrf2 in PANC-1 cells that induces antiproliferative
and apoptotic response possibly by attenuating NF-κB
[78-80]
Scopoletin
7-Hydroxy-6-methoxy-2H-chromen-2-one
Scopoletin inhibited the growth of all the cell lines and the IC50 ranged between 7.5 to 25µM.
Scopoletin exerted its anticancer effects on human cervical cancer cell lines by triggering apop-
tosis, cell cycle arrest, inhibition of cell invasion and PI3K/AKT signaling pathway.
Scopoletin caused significant suppression of sprouting of micro vessels in rat aortic explants
with IC50 (median inhibitory concentration) 0.06μM. Scopoletin (100 and 200mg/kg) strongly
inhibited (59.72 and 89.4%, respectively) vascularization in matrigel plugs implanted in nude
mice. In the tumor xenograft model, scopoletin showed remarkable inhibition on tumor growth
(34.2 and 94.7% at 100 and 200mg/kg, respectively).
[81, 82]
Angelicin
Furo[2,3-h]chromen-2-one
In human SH-SY5Y neuroblastoma cells, angelicin increased cellular cytotoxicity in a dose-
and time-dependent manner with IC50 of 49.56μM at 48h of incubation. Angelicin dose-
dependently down-regulated the expression of anti-apoptotic proteins including Bcl-2, Bcl-xL
and Mcl-1.
[83]
Bergamottin
4-[(2E)-3,7-dimethylocta-2,6-dienoxy]furo[3,2-g]chromen-
7-one
Bergamottin has potent antiproliferative effects on the A549 cells. Bergamottin shows both
concentration-dependent as well as time-dependent growth inhibitory effects against these cells.
The anticancer efficacy of Bergamottin under in vivo conditions using female BALB/c nude
mice was determined and in xenograft NSCLC model it reduced the tumor size significantly.
[84]
(Table 3) contd….
Nanoformulations of Coumarins and the Hybrid Molecules of Co umarins Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 1803
Name and Chemical Structure of Coumarins Pharmacological Activity Refs.
Limettin
(Citropten)
5,7-dimethoxychromen-2-one
Citropten conjugated with NDs showed antimitotic properties in B16F10 cells. [85]
Methoxalen
9-methoxy-7H-furo(3,2-g)chromen-7-one
Microemulsions vs. chitosan derivative-coated microemulsions for dermal delivery of 8-methoxypsoralen
(8-MOP) was investigated and the dermal delivery of microemulsion of 8-MOP was more efficient, which
might also be important for phototherapy applications in dermal carcinomas.
[86]
Aurapten
7-((E)-3, 7-Dimethylocta-2, 6-dienyloxy)-2H-
chromen-2-one
Aurapten decreases the secretion of Matrix Metalloproteinase-2 (MMP-2) as well as key inflammatory
mediators, including IL-6, IL-8, and chemokine (C-C motif) ligand-5(CCL5)
[87]
İsopimpinellin
4,9-Dimethoxy-7H-furo[3,2-g]chromen-7-one
Isopimipnellin inhibited the growth of human bladder carcinoma cell line [88]
Marmesin
O O
O
H
H
3
C
CH
3
HO
(2S)-2-(2-hyd roxypropan-2-y l)-2,3-d ihydrofuro [3,2-
g]chromen-7-one
Cytotoxic effects against the human leukemia (U937) cell line with IC50 of 40µM
Lower cytotoxic effects against normal human monocytes with IC50 of 125µM
Increase in the Bax/Bcl-2 ratio through the upregulation of Bax and downregulation of Bcl-2
Induce ROS mediated alterations in mitochondrial membrane potential, G2/M cell cycle arrest and signifi-
cantly inhibited leukemia cell migration potential
[89]
Rutamarin
O
OO
CH
3
CH
3
OO
CH
3
H
3
C
CH
3
CH
2
2-[6-(2-methylbut-3-en-2-yl)-7-oxo-2,3-
dihydrofuro[3,2-g]chromen-2-yl]propan-2-yl acetate
Extraction of Rutamarin from Ruta angustifolia Pers. and evaluated the cytotoxic activity against two
human carcinoma cell lines (HT29) and a normal human colon fibroblast cell line (CCD-18Co) using sul-
forhodamine B cytotoxicity assay.
Rutamarin exhibited remarkable cytotoxic activity against HT29 cells (IC50 value of 5.6μM) but was not
toxic to CCD-18Co cells.
[90]
Chalepin
O
OO
CH
3
CH
3
HO
H
3
C
CH
3
CH
2
2-(2-hydroxypropan-2-yl)-6-(2-methylbut-3-en-2-
yl)-2,3-dihydrofuro[3,2-g]chromen-7-one
Chloroform extract of Ruta angustifolia showed higher cytotoxic activity against MCF7, HT29, HCT116,
but mild cytotoxicity against MDA-MB231 and no activity against MCR5 (IC50>100μg/mL).
Chalepin and rutamarin isolated from Ruta angustifolia. Chalepin displayed remarkable cytotoxicity against
all tested cancer cell lines but no activity against MCR5. Rutamarin, on the other hand, showed remarkable
cytotoxicity activity against MCF7, HT29, and HCT116, whereas no activity against MDA-MB-231 and
MRC5.
Chalepin exhibited excellent cytotoxicity against A549 cell line with an IC50 value of 8.69±2.43μg/ml
(27.64μM).
A549 cells were treated with 36μg/ml of chalepin caused the decrease of anti-apoptotic proteins Bcl-2,
survivin, XIAP, Bcl-XL, cFLIP in a time-dependent manner and also level of PARP
Chalepin isolated from the chloroform extract Ruta angustifolia L and tested its ability to inhibit molecular
pathways of human lung carcinoma (A549) cells.
It causes S phase cell cycle arrest, NF-κB pathway inhibition, and STAT-3 inhibition, induces an extrinsic
apoptotic pathway.
Cytotoxic activity against MCF7 cells, whereas, it shows moderate cytotoxic activity against MDA-MB231
cells. Interestingly, chalepin did not present any toxicity against MRC5 normal cell line
Displays typical apoptotic features such as membrane blebbing, DNA fragmentation, chromatin condensa-
tion and apoptotic bodies' formation following chalepin treatment against MCF7 cells at different concen-
tration for 48h.
The protein expressions of caspases-8, -9 and cleaved PARP1 were upregulated, which correlated well with
increased caspase-3 activity.
[91-
94]
(Table 3) contd….
1804 Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 Yerer et al.
Name and Chemical Structure of Coumarins Pharmacological Activity Refs.
Columbianadin
O
OO
O
CH3
CH3
O
H3C
H3C
2-[(8S)-2-oxo-8,9-dihydrofuro[2,3-h]chromen-8-
yl]propan-2-yl (Z)-2-methylbut-2-enoate
Columbianadin isolated from Angelica decursiva (Umbelliferae) and evaluated anticancer activity
against HCT-116 human colon cancer cells.
At low concentration (25μM), it induced apoptosis (modulation of caspase-9, caspase-3, Bax, Bcl-
2, Bim and Bid) and high concentration (50μM), it induced necroptosis (RIP-3, and caspase-8).
It also causes the accumulation of ROS and imbalance in the intracellular antioxidant enzymes
such as SOD-1, SOD-2, catalase and GPx-1.
[95]
Nodakenetin
O
OO
H
3
C
H
3
C
HO
(2R)-2-(2-hydroxypropan-2-yl)-2,3-dihydrofuro[3,2-g]
chromen-7-one
Nodakenetin could significantly inhibit the proliferation of the leukemia HL-60 cells (IC50 of
16µM).
G2/M cell cycle arrest and induction of apoptosis, prompted mitochondrial apoptosis, alteration in
the apoptosis-related protein expression (Bax and Bcl-2).
[96]
(-)-deltoin
O O
O
CH
3
O
H
3
C
O
Isoxazolines and aziridines Derivatives of (-)-deltoin
O O
O
CH
3
O
H
3
C
O
N
OCH
3
R
H
3
C
OO
O
CH
3
O
H
3
C
O
N
CH
3
H
3
C
R
R= C
6
H
5
, 4-ClC
6
H
4
, 4-CH
3
C
6
H
4
R= C
6
H
5
, 2-ClC
6
H
3
N
(-)-deltoin exhibited good cytotoxic activity against the colon cell line HCT-116 (IC50 of
14.3±0.2µM) and a moderate cytotoxic activity against the ovary cell lines IGROV-1 and
OVCAR-3 with IC50 values of 49.0±2.1 and 49.0±1.8µM, respectively.
Phenyl substituted Isoxazolines derivative of (-)-deltoin is more potent than other with IC50 of
3.3±0.1µM (HCT-116), >100µM (IGROV-1), 63.0± 5.3µM (OVCAR-3) as well as the diastereoi-
someric mixture of 2-Chloropyridine substituted Isoxazolines derivative showed more potency
against HCT-116 with IC50 of 4.3±0.1.
Diastereoisomeric mixture of substituted phenyl aziridines derivative of (-)-deltoin showed more
potency against HCT-116 than the other two cell lines.
IC50 (µM) Phenyl group
HCT-116 IGROV-1 OVCAR-3
C6H5 5.9± 0.1 39.8 ±3.0 71.0± 2.6
4-ClC6H4 6.1± 0.7 31.6± 2.1 56.0± 2.9
4-CH3C6H4 7.3± 0.9 6.3± 0.5 18.0 ±0.7
[97]
(-)-5-hydroxyprantschimgin
O O
O
OH
O
H
3
CCH
3
O
CH
3
H
3
C
(-)-5-hydroxydeltoin
O O
O
OH
O
H
3
CCH
3
O
CH
3
H
3
C
(-)-5-hydroxyprantschimgin and (-)-5-hydroxydeltoin phytochemicals investigation of the roots of
Ferula lutea (Poir.) Maire and evaluation of cytotoxicity against human colorectal cancer cell lines.
The mixture of both compounds showed cytotoxic activity with IC50 values of 0.29±0.05 and
1.61±0.57µM against the cell lines HT-29 and HCT 116, respectively.
[98]
(Table 3) contd….
Nanoformulations of Coumarins and the Hybrid Molecules of Co umarins Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 1805
Name and Chemical Structure of Coumarins Pharmacological Activity Refs.
8-methoxysmyrindiol
OO
O
CH
3
H
3
C
HO
OH
OCH
3
8-methoxysmyrindiol extracted from Gerbera anandria (Compositae) and tested its cytotoxicity against
HeLa (human cervical cancer), MCF-7 (human breast cancer), HepG2 (Hepatocellular carcinoma),
HCT116 (human colon cancer), A549 (human lung adenocarcinoma), A375-S2 (human melanoma),
HT1080 (human fibrosarcoma), HL60 (myeloid leukemia).
8-methoxysmyrindiol showed more cytotoxicity against MCF-7, HCT11 and A549 cells with the highest
potency in HepG2 cells (IC50 = 5.3±0.2μg/ml).
[99]
Decursin
O OO
CH
3
H
3
C
O
OCH
3
H
3
C
Decursinol Angelate
O OO
CH
3
H
3
C
O
OCH
3
CH
3
Decursinol
O OO
CH
3
H
3
C
HO
4ʹʹ-hydroxytigloyldecursinol
O OO
CH
3
H
3
C
O
OHOH
2
C
CH
3
4ʹʹ-hydroxydecursin
O OO
CH
3
H
3
C
O
OCH
3
HOH
2
C
(2ʹʹR,3ʹʹR)-epoxyangeloyldecursinol
O OO
CH
3
H
3
C
O
O
CH
3
O
CH
3
(2ʹʹS,3ʹʹS)-epoxyangeloyldecursinol
O OO
CH
3
H
3
C
O
O
CH
3
O
CH
3
Ethanolic extract of Korean Angelica gigas Nakai majorly includes Decursin, Decursinol angelate, De-
cursinol and minor amount of other pyranocoumarins. In vivo as well as in vitro efficacy of this extract
was studied against bladder, breast, colon, lung and prostate cancers, sarcoma, myeloma and leukemia.
Angelica gigas Nakai (AGN) root ethanolic extract starting 8 weeks of age inhibited the growth of prostate
epithelium and Neuroendocrine carcinomas (NE-Ca) in the Transgenic Adenocarcinoma of Mouse Pros-
tate (TRAMP) model.
The growth of TRAMP Dorsolateral Prostate (DLP) in Angelica gigas Nakai and decursin with its isomer
decursinol angelate treated mice was inhibited by 66% and 61% at 16 weeks and by 67% and 72% at 28
weeks, respectively.
[100-
103]
Xanthyletin
OO O
H
3
C
H
3
C
Xanthoxyletin was isolated from Erythrina variegate and antiproliferative effects of Xanthoxyletin were
studied against SGC-7901 cells
induce apoptosis and cell cycle arrest, DNA damage and apoptosis through mitochondrial dysfunction, and
cell cycle arrest at S phase in a dose-dependent manner and production of reactive oxygen species in SGC-
7901 cells
Xanthoxyletin arrested the cell cycle of SGC-7901 cells at the S phase. The percentages of cells were
increased from 25.58% (in untreated cells) to 37.52% and 58.61% in cells treated with 200 and 400µM of
Xanthoxyletin respectively for 48h.
Xanthoxyletin induced apoptosis in SGC-7901 cells and the percentage of apoptotic cells was 41.47%
after the exposure of 400µM of Xanthoxyletin for 48 h as compared to 2.6% in the control group.
The fluorescence intensity was 97.30%, 70.09%, and 53.17% after treatment with 0, 200, and 400µM of
Xanthoxyletin respectively for 48h
[104]
(Table 3) contd….
1806 Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 Yerer et al.
Name and Chemical Structure of Coumarins Pharmacological Activity Refs.
Angular-type pyranocoumarin
OO O
H
3
C
H
3
COR
1
OR
2
R
1
OCH
3
CH
3
R
2
OCH
3
CH
3
OO
CH
3
OO O
H
3
C
H
3
COR
1
OR
2
R
1
R
2
O
CH
3
O
CH
3
H
3
C
O
CH
3
O
H
3
CCH
3
O
CH
3
O
CH
3
Isolation of new Angular-type pyranocoumarin from the roots of Peucedanum praeruptorum and evalua-
tion of the Multidrug-Resistance (MDR) reversal in human sarcoma MES-SA/Dx5 and Lipopolysaccha-
ride (LPS)-induced RAW 264.7 cells.
These inhibited the efflux of drugs by MDR protein, indicating the reversal of MDR and became an effec-
tive candidate for MDR forms of cancer.
[105]
Praeruptorin A
O OO
H
3
C
H
3
C
OO
CH
3
CH
3
O
CH
3
O
Significantly inhibited cell proliferation, colony formation, migration, invasion, and wound closure of
HeLa and SiHa cells
Induced cell cycle arrest at G0/G1 phase
Upregulated Rb, p16, p21 and p27 proteins
Downregulated cyclin D1 and S-phase kinase-associated protein 2 (Skp2) proteins.
Reduced expression of Matrix Metalloproteinase-2 (MMP-2) and increased expression of Tissue Inhibitor
of Metalloproteinase-2 (TIMP-2)
Suppressed ERK1/2 activation
Increased the effect of PD98059 (a specific MEK1/2 inhibitor) in downregulation of MMP-2 and upregu-
lation of TIMP-2
Inhibited the effect of 12-O-tetradecanoylphorbol-13-acetate (TPA) on upregulation of ERK1/2 activation,
MMP-2 expression, cellular migration, and invasion of HeLa cells.
[106]
(±) Praeruptorin A
O OO
H
3
C
H
3
C
OO
CH
3
CH
3
O
CH
3
O
(±) Praeruptorin B
OO
O
H
3
C
H
3
C
OO
CH
3
CH
3
O
O
CH
3
H
3
C
Methanolic extract of Peucedanum praeruptorum DUNN contains mainly (±) praeruptorin A and (±)
praeruptorin B
At 300µg/ml, PPME inhibited cell growth by 51.2% (P<0.01), probably linked to the high concentration
of PA and PB and exhibited antiproliferative and cytotoxic activities on the SGC7901 cells (gastric cancer
cells)
Enhanced the actions of doxorubicin on SGC7901 cells and reduced the dose of DOX for the desired
effects.
[107]
(Table 3) contd….
Nanoformulations of Coumarins and the Hybrid Molecules of Coumarins Anti-Cancer Agents in Medicinal C hemistry, 2020, Vol. 20, No. 15 1807
Name and Chemical Structure of Coumarins Pharmacological Activity Refs.
(+)-Praeruptorin A
O OO
H3C
H3C
OO
CH3
CH3
O
CH3
O
(+)-Praeruptoirn B
O OO
H
3
C
H
3
C
OO
CH
3
CH
3
O
O
CH
3
H
3
C
(+)-Praeruptoirn E
O OO
H
3
C
H
3
C
OO
CH
3
CH
3
O
O
CH
3
H
3
C
Ethanolic extract two-herb medicine formula consisting of Descurainiae sophia Semen and Peucedani
praeruptorum Radix
It consists of (+)-Praeruptoirn A (10.88mg/g), (+)-Praeruptoirn B (3.37mg/g), (+)-Praeruptoirn E
(1.32mg/g), Isoquercitrin (1.17mg/g), and Isorhamnetin-3-o-β-glucopyranoside (0.91mg/g)
The combination treatment of this extract with oxaliplatin or irinotecan remarkably enhanced the antitumor
activity of each anti-cancer drug and delayed tumor growth by 47.1-74.6% or by 74.4-82.9%, respectively
The Ki-67 expression for tumor cell proliferation and the CD31 for angiogenesis were decreased, and
tunnel staining for tumor cell apoptosis was remarkably increased.
[108]
Anomalin
O OO
O
O
CH
3
H
3
C
H
3
C
H
3
CO
O
CH
3
CH
3
Isolation of Anomalin from the roots of Peucedanum japonicum Thunb. (Umbelliferae) and evaluate its
efficacy against the human cancer (HeLa, HepG2, SNU-16, and AGS) cell lines.
Anomalin showed most potent cytotoxic activity against SNU-16 cells (IC50 of 18.24μg/mL) with no sig-
nificant cell death observed in the CCD-25Lu human normal lung fibroblast cells.
[109]
cis-khellactone
OO
O
H
3
C
H
3
COH
OH
Cytotoxic activities of cis-khellactone were evaluated on the proliferation and viability of MCF7 and
MDA-MB-231 human breast cancer and MCF10A normal cell lines and showed three types of pro-
grammed cell death that is apoptosis, autophagy-mediated cell death, and necrosis/necroptosis
The cis-khellactone at 10 or 20μg/ml concentration showed the best induction of cell death in the MCF7
and MDA-MB-231 cells, while normal cell is much less sensitive than cancer cell lines.
[110]
Dentatin
OO O
OCH
3
CH
3
CH
3
H
2
C
H
3
C
H
3
C
Antiproliferative potential of dentatin (a natural coumarin isolated from Clausena excavata Burm. F)
against prostate cancer
Treatment with dentatin dose-dependently inhibited cell growth of PC-3 and LNCaP prostate cancer cell
lines, whereas it showed less cytotoxic effects on the normal prostate epithelial cell line (RWPE-1)
Downregulated expression levels of antiapoptotic molecules (Bcl-2, Bcl-xl, and Survivin)
Disruption of mitochondrial membrane potential, cell membrane permeability, and release of cytochrome c
from the mitochondria into the cytosol
Inhibited TNF-α-induced nuclear translocation of p65, suggesting dentatin as a potential NF-κB inhibitor
Strong cytotoxicity against an activity with IC50 ranging from 5-10µg/ml against human promyelocytic
leukemia (HL-60), human breast cancer (MCF-7), human cervical cancer (HeLa), and human colon cancer
(HT-29) cells
Dentatin showed the IC50 values at 24, 48 or 72 hours against PC-3 cells (9.60±1.53μM, 8.15±2.41μM,
3.47±0.58μM, respectively); whereas against LNCaP cella (7.32±2.56μM, 6.79±1.90μM, 3.76±1.21μM),
respectively. It showed a less cytotoxic effect on RWPE-1 normal prostate epithelial cell line with IC50>
20μM at 24, 48 or 72 hours.
[111,
112]
(Table 3) contd….
1808 Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 Yerer et al.
Name and Chemical Structure of Coumarins Pharmacological Activity Refs.
Xanthoxyletin
O O O
H
3
C
H
3
C
OCH
3
Braylin
OO O
H
3
CO
H
3
C
H
3
C
6-methoxyseselin
OO O
H
3
CO
H
3
C
H
3
C
Xanthyletin
OO O
H
3
C
H
3
C
Xanthoxyletin, braylin, 6-methoxyseselin and other coumarins from ethanol extract of bark of Juglans
mandshurica and evaluated in vitro cytotoxicity against two liver cancer cell lines (HepG2 and Hep3B)
Xanthoxyletin and 6-methoxyseselin induced apoptosis and showed moderate antitumor activity against
both cell lines with IC50 values for HepG2 (62.30μM, 76.12μM) and Hep3B (66.53μM, 66.14μM) for 48h,
respectively.
Braylin and Xanthyletin showed cytotoxicities against Hep3B cell line with IC50 of 74.08 and 62.86μM,
respectively.
[113]
3',4'-Disenecioylkhellactone
O OO
H
3
C
H
3
CO
O
O
H
3
C
CH
3
CH
3
H
3
C
O
3′,4′-Disenecioylkhellactone strongly suppressed cell proliferation and induced caspase-mediated apoptosis
in AGS human gastric cancer cells.
It effectively suppressed signal transducer and activator of transcription 3 (STAT3) tyrosine phosphoryla-
tion, decreased STAT3 translocation to the nucleus and expression of STAT3 target genes.
[114]
visnadin
O OO
H
3
C
H
3
CO
H
3
C
O
O
O
CH
3
CH
3
The petroleum ether and CHCl3 fractions of Ammi visnaga L. flowering aerial parts consisting of khellin,
visnagin (furanochromones) and visnadin (pyranocoumarins) showed the most significant cytotoxic activ-
ity against malignant melanoma (HT 144) cell lines with an IC50 value of 0.400mg/ml using doxorubicin as
a reference anticancer drug.
[115]
O OO
H
3
C
H
3
C
OR
OR
OO
H
3
CO
H
3
CO
O
H
3
CO
H
3
CO
H
3
CO
O
R=
7,8-pyranocoumarins exhibited activity in modulating Pgp. At 4µM achieved 91%~99% decrease in IC50
value of anticancer agent’s vinblastine, doxorubicin, puromycin and paclitaxel.
Pgp-overexpressing human tumor cell lines HepG2/Dox (hepatocarcinoma), K562/Dox (leukemia) and KB
V1 (epidermoid carcinoma).
[116]
OO O
OH
R
CH
3
H
3
C
H
3
C
R= C
2
H
5
, C
3
H
7
, C
6
H
13
,
C
7
H
15
, C
10
H
21
3-alkyl,4- methylpyranocoumarins inhibited the cell proliferation of MDA-MB-468 and SK-OV-3 cells by
53-74%.
[117]
(Table 3) contd….
Nanoformulations of Coumarins and the Hybrid Molecules of Co umarins Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 1809
Name and Chemical Structure of Coumarins Pharmacological Activity Refs.
O OO
H
3
C
H
3
CO
O
CH
3
O
O
4-methyl-(3'S,4'S)-cis-khellactone derivatives were asymmetrically synthesized and evaluated
with respect to cytotoxic activity through MTT assay against HepG-2 (human liver carci-
noma), SGC-7901 (human gastric carcinoma), LS174T (human colon carcinoma) cell lines.
Among these, tigloyl derivative was the most potent and exhibited strong cytotoxicity, with
IC50 values ranging from 8.51 to 29.65μM.
[118]
Alloxanthoxyletin fatty acids esters
O O
CH
3
O
H
3
CCH
3
OR
O
Synthesis of alloxanthoxyletin fatty acids esters and evaluation of in vitro anticancer activity
against HTB-140 and A549 cancer cells using cisplatin doxorubicin as reference drugs.
Among these, Oleic (R=C17H33), α - linolenic acid (R= C17H29), Conjugated linoleic acid
(R=C17H31) and Docosahexaenoic acid (R=C21H31) derivatives showed a stronger inhibi-
tory effects on cancer cells (HTB-140 and A549) than in normal cells (HaCaT).
These derivatives possessed the highest cytotoxic potential against HTB-140 cells with IC50
of 14.4-35.4μM.
These agents showed statistically significant effect against both tumor cell lines when oleic
derivative used in the concentration of 40μM, linolenic derivatives in concentration of 20μM
and docosahexaenoic acid at 10μM concentration.
[119]
Alloxanthoxyletin O-aminoalkyl derivatives
O O
CH
3
O
H
3
CCH
3
OR
Seselin O-aminoalkyl derivatives
OO
CH
3
O
H
3
C
H
3
C
OR
Synthesis of O-aminoalkyl substituted alloxanthoxyletins and seselins and evaluated for their
anticancer toxicity against HaCaT, A549 and HTB-140 cells lines.
Seselin and alloxanthoxylein derivatives increase the sensitivity of tumor cells to drug-
induced cell death as well as tumor cells showed a higher sensitivity to tested compounds than
normal cells.
Diethylamine, dimethyl amine and piperidine Alloxanthoxyletin derivatives showed the
highest cytotoxic potential against HTB-140 cells with IC50 of 2.48, 2.80 and 2.98μM, re-
spectively.
[120]
Gemcitabine−Coumarin−Biotin conjugate
OOO
OO SSO
O
OO
OH FF
N
N
O
NH
2
NN
H
NH
NH
O
S
NH
NH
O
The active molecule is preferentially taken up by A549 cells [56]
Umbelliprenin
O
O
O
The cytotoxic effect of umbelliprenin (7-prenyloxycoumarins) was discovered on LLC cells
and mouse splenocytes
[57]
Coumarin-Chlorambucil Conjugate
O
O
OH
O
ONCl
Cl
Cell viability was evaluated utilizing MTT assay in breast cancer cells MDA-MB-231 [58]
(Table 3) contd….
1810 Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 Yerer et al.
Name and Chemical Structure of Coumarins Pharmacological Activity Refs.
AD-013
OO
O
O
Compound showed some selectivity against MCF-7 cancer cells as compared with MCF-
10A healthy cells
[59]
OOR
O
These molecules decreased the proliferation of PC3 cells [60]
7-(benzyloxy)-4-(hydroxymethyl)-2H-1-ben zopyran -2-one
OOO
OH
Anticancer effects of intracellular self-assembly started by light. CCK-8 assay quantified
the viability of both HepG2 and HeLa cells
[61]
OOO
NH
O
Drug delivery of 7-ethyl-10-hydroxy camptothecin (SN-38), two nanoparticles (P-II and
P-IV micelles) were viewed and chosen for the cure of colon cancers.
[63]
Farnesiferol C
OOO
O
The anticancer impact of Dendrosomal Farnesiferol C (DFC) on the AGS gastric cancer
cell line.
[65]
4-methyl-7-hydroxy coumarin
OOOH
Encapsulated 4-methyl-7-hydroxy coumarin (SC), to test if nano-encapsulated SC can be
a more efficient anti-cancer agent.
[66]
Matrix metalloproteinase-2 (MMP-2) probe
OON
O
NH
O
R
For the detection of cervical cancer, it ensures the probability based on MMP-2 [68]
Squaric acid-Coumarin-Chlorambucil
(Sq-Cou-Cbl) nanoconjugates
O
O
O
NH
O
O
OH NCl
Cl
( )
On HeLa cells in vitro works demonstrated that Sq-Cou-Cbl nanoconjugates showed an
advanced anticancer activity
[70]
(Table 3) contd….
Nanoformulations of Coumarins and the Hybrid Molecules of Co umarins Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 1811
Name and Chemical Structure of Coumarins Pharmacological Activity Refs.
7-hydroxy-6-methoxy-2H-chromen-2-one (Scopoletin);
O O
H
3
CO
HO
3-acrylamido-6-methoxy-2-oxo-2H-chromen-7-yl acetate (7a);
O O
H
3
CO
AcO
H
N
O
N-(7-hydroxy-6-methoxy-2-oxo-2H-chromen-3-yl)acrylamide (7b);
O O
H
3
CO
HO
H
N
O
N-(7-hydroxy-6-methoxy-2-oxo-2H-chromen-3-yl)-2-(piperidin-1-
yl)acetamide (7e);
O O
H
3
CO
HO
H
N
O
N
N-(7-hydroxy-6-methoxy-2-oxo-2H-chromen-3-yl)-2-(4-methylpiperidin-
1-yl)acetamide (7f);
O O
H
3
CO
HO
H
N
O
N
3-(2-chloroacetamido)-6-methoxy-2-oxo-2H-chromen-7-yl acetate (8a);
O O
H3CO
AcO
H
N
O
Cl
6-methoxy-3-(2-(4-methylpiperazin-1-yl)acetamido)-2-oxo-2H-chromen-
7-yl acetate (8e);
O O
H3CO
AcO
H
N
O
N
N
Scopoletin derivatives were fabricated from the reaction of β-aminopropamide, α-
aminoacetamide, acrylamide, and pure scopoletin and carried out in vitro cyto-
toxic activities by MTT.
IC50 (µM)
Molecules
A549 MDA-MB-
231 HEPG2
Scopoletin >100 >100 >100
7a >100 8.882±0.93 8.710±0.94
7b >100 21.96±2.29 4.80±0.11
7e >100 25.46±1.42 58.20±2.16
7f >100 8.579±0.48 14.15±0.71
8a 3.961±0.89 2.195±0.21 3.721±0.61
8e 41.16±1.73 71.86±2.45 30.97±1.49
[121]
attention. Ganghopadyay et al. made use of a multi-arm 15 PEG,
functionalized with a targeting unit biotin and a coumarin fluoro-
phore for site-specific and image guided synergic treatment of can-
cer cells. UV/vis light (≥365nm) with a moderate quantum yield of
~0.37, anticancer drug chlorambucil was released by coumarin
chromophore in a photo-controlled way. In addition to that, cou-
marin created singlet oxygen upon irradiation. In vitro application
of prepared organic polymeric nanoparticles (PEG-Bio-Cou-Cbl) in
Hela cell line demonstrated a reduction in cell viability of up to
~5% in the case of combined treatment of PDT and chemotherapy
while analogous organic polymeric NPs without the chemothera-
peutic drug (PEG-Bio-Cou) resulted in ~49% cell viability by PDT
protocol only [67].
The development of new strategies with high sensibility and
specificity is needed to improve the therapeutic rate of cervical
cancer. In normal cervical epithelial cells, Matrix Metalloprotein-
ase-2 (MMP-2) is low or absent, whereas it is overexpressed in
cervical intraepithelial neoplasia and cervical cancer. This provides
the possibility for the detection of cervical cancer based on MMP-2.
Connecting a fluorophore with a quencher via a MMP-2 substrate
peptide, a novel coumarin-based fluorescence resonance energy
transfer probe targeting MMP-2 was prepared to investigate cervi-
cal cancer by cell imaging. This MMP-2 probe potentially ensures a
specific and sensitive optic method for cervical cancer screening,
prognostic and diagnostic evaluation [68].
Nanogels with coumarin as a photocleavable group were de-
scribed as promising drug delivery systems in order to improve
anticancer treatment with hydrophobic drugs. Multi-stimuli respon-
sive nanogels based on biocompatible hydrophilic polymers have
been shown to increase circulating-time in the bloodstream, tumor-
targeting and reduction of systemic toxicity. Stefano et al. studied
the synthesis, characterization and biological perspectives of light-
and thermoresponsive Hyaluronic Acid (HA)-based nanogels con-
taining coumarin as photocleavable group. These gel particles were
obtained by temperature-induced self-assembly of dual stimuli
(light and temperature)-responsive HA derivatives. On the polysac-
charide backbone, it was prepared by grafting DEGMA and CMA-
based copolymer chains. In vitro biological studies showed that the
nanogels and their hydrophobic load are internalized by HeLa cells
(cervical cancer) in a concentration-dependent manner. No more
circulating nanogels were explored 24 h-post administration. A
hydrophobic dye accumulation was verified in tumoral tissues near
the blood vessels [69].
Chadhuri et al. improved photoresponsive fluorescent organic
nanoconjugates based on a single-component system by a synergis-
tic combination of Photodynamic Therapy (PDT) and chemother-
apy in order to efficiently cure cancer. They synthesized single-
component system by coupling the squaric acid and the coumarin-
chlorambucil conjugate. Multifunctional squaric acid-coumarin-
chlorambucil (Sq-Cou-Cbl) nanoconjugates were prepared by
“simple reprecipitation technique”. On HeLa cells, in vitro works
demonstrated that Sq-Cou-Cbl nanoconjugates showed an advanced
anticancer activity by the synergistic cytotoxic impact of PDT and
chemotherapy [70].
1812 Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 Yerer et al.
1.2.10. Fibrosarcoma
In nanomedicine, the potential applications of Fe3O4 Magnetite
Nanoparticles (MNPs) are well known as drug delivery systems.
Khorramizadeh et a l. prepared umbelliprenin-coated Fe3O4 MNPs.
The antiproliferative effect of the combination was evaluated in
vitro. Using transmission electron microscopy, energy-dispersive
spectroscopy, and X-ray diffraction spectroscopy techniques, Fe3O4
MNPs were characterized after synthesis. Umbelliprenin (the natu-
ral candidate coumarin compound) was isolated and identified.
Umbelliprenin-coated Fe3O4 MNPs were prepared, utilizing pre-
cipitation method. In vitro, HT-1080 cells (fibrosarcoma) were
cultured and MTT assay was carried out for evaluating the possible
cytotoxic effects of umbelliprenin-coated Fe3O4 MNPs in viable
cells. The results showed that umbelliprenin had middle antiprolif-
erative effects with IC50 value of 50µg/ml. The combination of
umbelliprenin and Fe3O4 MNPs demonstrated the IC50 value of
9µg/ml [71].
Taking into account the anticancer effects of coumarin deriva-
tives and hybrid molecules and the nanoformulations, these com-
pounds are summarized in Table 3 [72-121].
CONCLUSION
In this review article, antitumor activities of nanoformulations
of different coumarins are thoroughly described in diverse experi-
mental models of both solid tumors and hematological malignan-
cies. These data clearly demonstrate the power of joining the poten-
tial anticancer properties of natural plant compounds and novel
technological advances to hopefully achieve safe and efficient mo-
dalities for the treatment of malignant disorders. In fact, there are
numerous data available about the in vitro anticancer properties of
structurally diverse coumarins. However, efforts to translate these
findings to the human systems have mostly failed due to the issues
of bioavailability and biotransformation of natural polyphenolic
compounds. As shown in this review, nanotechnology might be the
key to overcome these obstacles, improving the delivery of coumar-
ins specifically to cancerous sites and thereby facilitating the im-
plementation of anticancer potential of these plant-derived agents.
Nevertheless, despite the promising results described so far in
applying nanoformulations in preclinical studies, there are still sev-
eral aspects needed to unravel before achieving an ideal nanovehi-
cle for coumarins. The important factors at this probably involve
not only the specificity and efficiency of antican cer action, but also
the long-term safety and cost-effectiveness. Therefore, besides the
continuation of intense preclinical studies for identification of the
best composition of coumarins-loaded nanoformulations, clinical
trials should also be designed in the future, to comprehend the real
potential of nanoformulations of coumarins in controlling cancer
development, progression and spread.
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
None.
CONFLICT OF INTEREST
The authors confirm that this article content has no conflict of
interest.
ACKNOWLEDGEMENTS
Declared none.
REFERENCES
[1] Nurgali, K.; Jagoe, R.T.; Abalo, R. Adverse effects of cancer che-
motherapy: Anything new to improve tolerance and reduce seque-
lae? Front. Pharmacol., 2018, 9, 245.
http://dx.doi.org/10.3389/fphar.2018.00245 PMID: 29623040
[2] Rayan, A.; Raiyn, J.; Falah, M. Nature is the best source of anti-
cancer drugs: Indexing natural products for their anticancer bioac-
tivity. PLoS One, 2017, 12(11), e0187925.
http://dx.doi.org/10.1371/journal.pone.0187925 PMID: 29121120
[3] Seca, A.M.L.; Pinto, D.C.G.A. Plant secondary metabolites as
anticancer agents: successes in clinical trials and therapeutic appli-
cation. Int. J. Mol. Sci., 2018, 19(1), 263.
http://dx.doi.org/10.3390/ijms19010263 PMID: 29337925
[4] Matos, M.J.; Santana, L.; Uriarte, E.; Abreu, O.A.; Molina, E.;
Yordi, E.G. Coumarins-an important class of phytochemicals. In:
Phytochemicals-Isolation, Characterisation and Role in Human
Health; Rao, V., Ed.; InTech: UK, 2015; pp. 113-140.
[5] Jain, P.; Joshi, H. Coumarin: Chemical and pharmacological pro-
file. J. Appl. Pharm. Sci., 2012, 2(6), 236-240.
[6] Kesarwani, K.; Gupta, R.; Mukerjee, A. Bioavailability enhancers
of herbal origin: An overview. Asian Pac. J. Trop. Biomed., 2013,
3(4), 253-266.
http://dx.doi.org/10.1016/S2221-1691(13)60060-X PMID:
23620848
[7] Kallitsakis, M.G.; Carotti, A.; Catto, M.; Peperidou, A.; Hadjipav-
lou-Litina, D.J.; Litinas, K.E. Synthesis and biological evaluation
of novel hybrid molecules containing purine, coumarin and isoxa-
zoline or isoxazole moieties. Open Med. Chem. J., 2017, 11, 196-
211.
http://dx.doi.org/10.2174/1874104501711010196 PMID: 29387274
[8] Kahveci, B.; Yılmaz, F.; Menteşe, E.; Ülker, S. Design, synthesis,
and biological evaluation of coumarin–triazole hybrid molecules as
potential antitumor and pancreatic lipase agents. Arch. Pharm.
(Weinheim), 2017, 350(8), 1600369.
http://dx.doi.org/10.1002/ardp.201600369 PMID: 28543820
[9] Singh, H.; Singh, J.V.; Gupta, M.K.; Saxena, A.K.; Sharma, S.;
Nepali, K.; Bedi, P.M.S. Triazole tethered isatin-coumarin based
molecular hybrids as novel antitubulin agents: Design, synthesis,
biological investigation and docking studies. Bioorg. Med. Chem.
Lett., 2017, 27(17), 3974-3979.
http://dx.doi.org/10.1016/j.bmcl.2017.07.069 PMID: 28797799
[10] Thakur, A.; Singla, R.; Jaitak, V. Coumarins as anticancer agents: a
review on synthetic strategies, mechanism of action and SAR stud-
ies. Eur. J. Med. Chem., 2015, 101, 476-495.
http://dx.doi.org/10.1016/j.ejmech.2015.07.010 PMID: 26188907
[11] Teiten, M-H.; Dicato, M.; Diederich, M. Hybrid curcumin com-
pounds: a new strategy for cancer treatment. Molecules, 2014,
19(12), 20839-20863.
http://dx.doi.org/10.3390/molecules191220839 PMID: 25514225
[12] Piekuś-Słomka, N.; Mikstacka, R.; Ronowicz, J.; Sobiak, S. Hybrid
cis-stilbene molecules: Novel anticancer agents. Int. J. Mol. Sci.,
2019, 20(6), 1300.
http://dx.doi.org/10.3390/ijms20061300 PMID: 30875859
[13] Ansari, S.H.; Islam, F.; Sameem, M. Influence of nanotechnology
on herbal drugs: A review. J. Adv. Pharm. Technol. Res., 2012,
3(3), 142-146.
http://dx.doi.org/10.4103/2231-4040.101006 PMID: 23057000
[14] Bonifácio, B.V.; Silva, P.B.; Ramos, M.A.; Negri, K.M.S.; Bauab,
T.M.; Chorilli, M. Nanotechnology-based drug delivery systems
and herbal medicines: A review. Int. J. Nanomedicine, 2014, 9, 1-
15. PMID: 24363556
[15] Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-
Torres, M.D.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.;
Swamy, M.K.; Sharma, S.; Habtemariam, S.; Shin, H.S. Nano
based drug delivery systems: Recent developments and future
prospects. J. Nanobiotechnology, 2018, 16(1), 71.
http://dx.doi.org/10.1186/s12951-018-0392-8 PMID: 30231877
[16] Sabt, A.; Abdelhafez, O.M.; El-Haggar, R.S.; Madkour, H.M.F.;
Eldehna, W.M.; El-Khrisy, E.E.A.M.; Abdel-Rahman, M.A.;
Rashed, L.A. Novel coumarin-6-sulfonamides as apoptotic anti-
proliferative agents: Synthesis, in vitro biological evaluation, and
QSAR studies. J. Enzyme Inhib. Med. Chem., 2018, 33(1), 1095-
1107. http://dx.doi.org/10.1080/14756366.2018.1477137 PMID:
29944015
Nanoformulations of Coumarins and the Hybrid Molecules of Co umarins Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 1813
[17] Chuang, J-Y.; Huang, Y-F.; Lu, H-F.; Ho, H-C.; Yang, J-S.; Li, T-
M.; Chang, N-W.; Chung, J-G. Coumarin induces cell cycle arrest
and apoptosis in human cervical cancer HeLa cells through a mito-
chondria- and caspase-3 dependent mechanism and NF-kappaB
down-regulation. In Vivo, 2007, 21(6), 1003-1009.
PMID: 18210747
[18] Kimura, Y.; Sumiyoshi, M. Antitumor and antimetastatic actions of
dihydroxycoumarins (esculetin or fraxetin) through the inhibition
of M2 macrophage differentiation in tumor-associated macro-
phages and/or G1 arrest in tumor cells. Eur. J. Pharmacol., 2015,
746, 115-125.
http://dx.doi.org/10.1016/j.ejphar.2014.10.048 PMID: 25445053
[19] Lee, S.; Sivakumar, K.; Shin, W-S.; Xie, F.; Wang, Q. Synthesis
and anti-angiogenesis activity of coumarin derivatives. Bioorg.
Med. Chem. Lett., 2006, 16(17), 4596-4599.
http://dx.doi.org/10.1016/j.bmcl.2006.06.007 PMID: 16793260
[20] Khan, S.; Shehzad, O.; Cheng, M-S.; Li, R-J.; Kim, Y.S. Pharma-
cological mechanism underlying anti-inflammatory properties of
two structurally divergent coumarins through the inhibition of pro-
inflammatory enzymes and cytokines. J. Inflamm. (Lond.), 2015,
12(1), 47.
http://dx.doi.org/10.1186/s12950-015-0087-y PMID: 26221081
[21] Musa, M.A.; Cooperwood, J.S.; Khan, M.O.F. A review of cou-
marin derivatives in pharmacotherapy of breast cancer. Curr. Med.
Chem., 2008, 15(26), 2664-2679.
http://dx.doi.org/10.2174/092986708786242877 PMID: 18991629
[22] Govindaiah, P.; Dumala, N.; Grover, P.; Jaya Prakash, M. Synthe-
sis and biological evaluation of novel 4,7-dihydroxycoumarin de-
rivatives as anticancer agents. Bioorg. Med. Chem. Lett., 2019,
29(14), 1819-1824.
http://dx.doi.org/10.1016/j.bmcl.2019.05.008 PMID: 31104996
[23] Bhagat, K.; Bhagat, J.; Gupta, M.K.; Singh, J.V.; Gulati, H.K.;
Singh, A.; Kaur, K.; Kaur, G.; Sharma, S.; Rana, A.; Singh, H.;
Sharma, S.; Singh Bedi, P.M. Design, synthesis, antimicrobial
evaluation, and molecular modeling studies of novel indolinedione-
coumarin molecular hybrids. ACS Omega, 2019, 4(5), 8720-8730.
http://dx.doi.org/10.1021/acsomega.8b02481 PMID: 31459961
[24] Goud, N.S.; Ghouse, M.S.; Vishnu, J.; Pranay, J.; Alvala, R.; Talla,
V.; Qureshi, I.A.; Alvala, M. Synthesis and biological evaluation of
novel heterocyclic imines linked coumarin- thiazole hybrids as
anticancer agents. Anticancer Agents Med. Chem., 2019, 19(4),
557-566.
http://dx.doi.org/10.2174/1871520619666190207140120 PMID:
30734685
[25] Cavalcanti, E.B.V.S.; Félix, M.B.; Scotti, L.; Scotti, M.T. Virtual
screening of natural products to select compounds with potential
anticancer activity. Anticancer Agents Med. Chem., 2019, 19(2),
154-171.
http://dx.doi.org/10.2174/1871520618666181119110934 PMID:
30451120
[26] Karataş, M.O.; Olgundeniz, B.; Günal, S.; Özdemir, İ.; Alıcı, B.;
Çetinkaya, E. Synthesis, characterization and antimicrobial activi-
ties of novel silver(I) complexes with coumarin substituted N-
heterocyclic carbene ligands. Bioorg. Med. Chem., 2016, 24(4),
643-650.
http://dx.doi.org/10.1016/j.bmc.2015.12.032 PMID: 26740157
[27] Lacy, A.; O’Kennedy, R. Studies on coumarins and coumarin-
related compounds to determine their therapeutic role in the treat-
ment of cancer. Curr. Pharm. Des., 2004, 10(30), 3797-3811.
http://dx.doi.org/10.2174/1381612043382693 PMID: 15579072
[28] Karataş, M.O.; Uslu, H.; Sarı, S.; Alagöz, M.A.; Karakurt, A.;
Alıcı, B.; Bilen, C.; Yavuz, E.; Gencer, N.; Arslan, O. Coumarin or
benzoxazinone based novel carbonic anhydrase inhibitors: Synthe-
sis, molecular docking and anticonvulsant studies. J. Enzyme Inhib.
Med. Chem., 2016, 31(5), 760-772.
http://dx.doi.org/10.3109/14756366.2015.1063624 PMID: 26207513
[29] Innocenti, A.; Durdagi, S.; Doostdar, N.; Strom, T.A.; Barron,
A.R.; Supuran, C.T. Nanoscale enzyme inhibitors: Fullerenes in-
hibit carbonic anhydrase by occluding the active site entrance.
Bioorg. Med. Chem., 2010, 18(8), 2822-2828.
http://dx.doi.org/10.1016/j.bmc.2010.03.026 PMID: 20363143
[30] Karatas, M.O.; Alici, B.; Cakir, U.; Cetinkaya, E.; Demir, D.; Er-
gün, A.; Gençer, N.; Arslan, O. Synthesis and carbonic anhydrase
inhibitory properties of novel coumarin derivatives. J. Enzyme In-
hib. Med. Chem., 2013, 28(2), 299-304.
http://dx.doi.org/10.3109/14756366.2012.677838 PMID: 22512727
[31] Solarova, Z.; Kello, M.; Hamul’akova, S.; Mirossay, L.; Solar, P.
Anti-cancer effect of tacrine-coumarin derivatives on diverse hu-
man and mouse cancer cell lines. Acta Chim. Slov., 2018, 65(4),
875-881.
http://dx.doi.org/10.17344/acsi.2018.4519
[32] Kostova, I.; Raleva, S.; Genova, P.; Argirova, R. Structure-activity
relationships of synthetic coumarins as HIV-1 inhibitors. Bioinorg.
Chem. Appl., 2006, 2006, Article ID 68274.
http://dx.doi.org/10.1155/BCA/2006/68274 PMID: 17497014
[33] Bedoya, L.M.; Beltrán, M.; Sancho, R.; Olmedo, D.A.; Sánchez-
Palomino, S.; del Olmo, E.; López-Pérez, J.L.; Muñoz, E.; San Fe-
liciano, A.; Alcamí, J. 4-Phenylcoumarins as HIV transcription in-
hibitors. Bioorg. Med. Chem. Lett., 2005, 15(20), 4447-4450.
http://dx.doi.org/10.1016/j.bmcl.2005.07.041 PMID: 16137881
[34] Thomas, V.; Giles, D.; Basavarajaswamy, G.P.M.; Das, A.K.;
Patel, A. Coumarin derivatives as anti-inflammatory and anticancer
agents. Anticancer Agents Med. Chem., 2017, 17(3), 415-423.
http://dx.doi.org/10.2174/1871520616666160902094739 PMID:
27592545
[35] Kasumbwe, K.; Venugopala, K.N.; Mohanlall, V.; Odhav, B. Syn-
thetic Mono/di-halogenated coumarin derivatives and their antican-
cer properties. Anticancer Agents Med. Chem., 2017, 17(2), 276-
285.
http://dx.doi.org/10.2174/1871520616666160926112508 PMID:
27671300
[36] Wang, Y.; Yan, W.; Chen, Q.; Huang, W.; Yang, Z.; Li, X.; Wang,
X. Inhibition viral RNP and anti-inflammatory activity of coumar-
ins against influenza virus. Biomed. Pharmacother., 2017, 87, 583-
588.
http://dx.doi.org/10.1016/j.biopha.2016.12.117 PMID: 28081470
[37] Liu, H.; Wang, L.; Gao, H.; Qi, H.; Gao, Q.; Zhang, C. Aggrega-
tion-induced enhanced electrochemiluminescence from organic
nanoparticles of donor-acceptor based coumarin derivatives. ACS
Appl. Mater. Interfaces, 2017, 9(51), 44324-44331.
http://dx.doi.org/10.1021/acsami.7b15434 PMID: 29171261
[38] Traven, V.F.; Cheptsov, D.A.; Solovjova, N.P.; Chibisova, T.A.;
Voronov, I.I.; Dolotov, S.M.; Ivanov, I.V. Photoinduced formation
of the laser dye coumarin 6 from its dihydro derivatives. Dyes Pig-
ments, 2017, 146, 159-168.
http://dx.doi.org/10.1016/j.dyepig.2017.07.001
[39] Karatas, M.O.; Di Giuseppe, A.; Passarelli, V.; Alici, B.; Perez-
Torrente, J.J.; Oro, L.A.; Ozdemir, I.; Castarlenas, R. Pentacoordi-
nated Rhodium(I) complexes supported by coumarin-
functionalized N-heterocyclic carbene ligands. Organometallics,
2018, 37(2), 191-202.
http://dx.doi.org/10.1021/acs.organomet.7b00750
[40] Liu, X.G.; Zhang, S.S.; Jiang, C.Y.; Wu, J.Q.; Li, Q.; Wang, H.
Cp*Co(III)-catalyzed annulations of 2-alkenylphenols with CO:
Mild access to coumarin derivatives. Org. Lett., 2015, 17(21),
5404-5407.
http://dx.doi.org/10.1021/acs.orglett.5b02728 PMID: 26451846
[41] Mahdaviani, P.; Bahadorikhalili, S.; Navaei-Nigjeh, M.; Vafaei,
S.Y.; Esfandyari-Manesh, M.; Abdolghaffari, A.H.; Daman, Z.;
Atyabi, F.; Ghahremani, M.H.; Amini, M.; Lavasanifar, A.; Dinar-
vand, R. Peptide functionalized poly ethylene glycol-poly caprolac-
tone nanomicelles for specific cabazitaxel delivery to metastatic
breast cancer cells. Mater. Sci. Eng. C, 2017, 80, 301-312.
http://dx.doi.org/10.1016/j.msec.2017.05.126 PMID: 28866169
[42] Abdul Manaf, S.A.; Hegde, G.; Mandal, U.K.; Wui, T.W.; Roy, P.
Functionalized carbon nano-scale drug delivery systems from
biowaste sago bark for cancer cell imaging. Curr. Drug Deliv.,
2017, 14(8), 1071-1077.
http://dx.doi.org/10.2174/1567201813666161017130612 PMID:
27745545
[43] Alam, N.; Qayum, A.; Kumar, A.; Khare, V.; Sharma, P.R.; An-
dotra, S.S.; Singh, S.K.; Koul, S.; Gupta, P.N. Improved efficacy of
cisplatin in combination with a nano-formulation of pentacyclic
triterpenediol. Mater. Sci. Eng. C, 2016, 68, 109-116.
http://dx.doi.org/10.1016/j.msec.2016.05.093 PMID: 27524002
[44] Tao, W.; Zeng, X.W.; Zhang, J.X.; Zhu, H.J.; Chang, D.F.; Zhang,
X.D.; Gao, Y.F.; Tang, J.; Huang, L.Q.; Mei, L. Synthesis of cholic
acid-core poly(epsilon-caprolactone-ran-lactide)-b-poly(ethylene
glycol) 1000 random copolymer as a chemotherapeutic nanocarrier
for liver cancer treatment. Biomater Sci-UK, 2014, 2(9), 1262-
1274.
http://dx.doi.org/10.1039/C4BM00134F
1814 Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 Yerer et al.
[45] Li, Z.; Liu, K.; Sun, P.; Mei, L.; Hao, T.; Tian, Y.; Tang, Z.; Li, L.;
Chen, D. Poly(D, L-lactide-co-glycolide)/montmorillonite nanopar-
ticles for improved oral delivery of exemestane. J. Microencapsul.,
2013, 30(5), 432-440.
http://dx.doi.org/10.3109/02652048.2012.746749 PMID: 23517067
[46] Wang, J.; Liu, W.; Tu, Q.; Wang, J.; Song, N.; Zhang, Y.; Nie, N.;
Wang, J. Folate-decorated hybrid polymeric nanoparticles for
chemically and physically combined paclitaxel loading and tar-
geted delivery. Biomacromolecules, 2011, 12(1), 228-234.
http://dx.doi.org/10.1021/bm101206g PMID: 21158381
[47] Paiva, A.M.; Pinto, R.A.; Teixeira, M.; Barbosa, C.M.; Lima, R.T.;
Vasconcelos, M.H.; Sousa, E.; Pinto, M. Development of noncyto-
toxic PLGA nanoparticles to improve the effect of a new inhibitor
of p53-MDM2 interaction. Int. J. Pharm., 2013, 454(1), 394-402.
http://dx.doi.org/10.1016/j.ijpharm.2013.07.017 PMID: 23856033
[48] Bazylińska, U.; Zieliński, W.; Kulbacka, J.; Samoć, M.; Wilk, K.A.
New diamidequat-type surfactants in fabrication of long-sustained
theranostic nanocapsules: Colloidal stability, drug delivery and bio-
imaging. Colloids Surf. B Biointerfaces, 2016, 137, 121-132.
http://dx.doi.org/10.1016/j.colsurfb.2015.06.043 PMID: 26164204
[49] Zhao, T.; Chen, H.; Dong, Y.; Zhang, J.; Huang, H.; Zhu, J.;
Zhang, W. Paclitaxel-loaded poly(glycolide-co-ε-caprolactone)-b-
D-α-tocopheryl polyethylene glycol 2000 succinate nanoparticles
for lung cancer therapy. Int. J. Nanomedicine, 2013, 8, 1947-1957.
PMID: 23696703
[50] Su, Y.; Hu, J.; Huang, Z.; Huang, Y.; Peng, B.; Xie, N.; Liu, H.
Paclitaxel-loaded star-shaped copolymer nanoparticles for en-
hanced malignant melanoma chemotherapy against multidrug resis-
tance. Drug Des. Devel. Ther., 2017, 11, 659-668.
http://dx.doi.org/10.2147/DDDT.S127328 PMID: 28293102
[51] Kushwah, V.; Katiyar, S.S.; Dora, C.P.; Kumar Agrawal, A.; Lam-
prou, D.A.; Gupta, R.C.; Jain, S. Co-delivery of docetaxel and
gemcitabine by anacardic acid modified self-assembled albumin
nanoparticles for effective breast cancer management. Acta Bioma-
ter., 2018, 73, 424-436.
http://dx.doi.org/10.1016/j.actbio.2018.03.057 PMID: 29649635
[52] Lv, J.; Qiao, W.; Li, Z. Vesicles from pH-regulated reversible
gemini amino-acid surfactants as nanocapsules for delivery. Col-
loids Surf. B Biointerfaces, 2016, 146, 523-531.
http://dx.doi.org/10.1016/j.colsurfb.2016.06.054 PMID: 27419647
[53] Catti, L.; Kishida, N.; Kai, T.; Akita, M.; Yoshizawa, M. Polyaro-
matic nanocapsules as photoresponsive hosts in water. Na t. C om-
mun., 2019, 10(1), 1948.
http://dx.doi.org/10.1038/s41467-019-09928-x PMID: 31019192
[54] Attia, M.F.; Anton, N.; Bouchaala, R.; Didier, P.; Arntz, Y.; Mes-
saddeq, N.; Klymchenko, A.S.; Mely, Y.; Vandamme, T.F. Func-
tionalization of nano-emulsions with an amino-silica shell at the
oil-water interface. RSC Adv., 2015, 5(91), 74353-74361.
http://dx.doi.org/10.1039/C5RA12676B
[55] Lee, J.H.; Kim, K.Y.; Jin, H.; Baek, Y.E.; Choi, Y.; Jung, S.H.;
Lee, S.S.; Bae, J.; Jung, J.H. Self-assembled coumarin nanoparticle
in aqueous solution as selective mitochondrial-targeting drug deliv-
ery system. ACS Appl. Mater. Interfaces, 2018, 10(4), 3380-3391.
http://dx.doi.org/10.1021/acsami.7b17711 PMID: 29302967
[56] Maiti, S.; Park, N.; Han, J.H.; Jeon, H.M.; Lee, J.H.; Bhuniya, S.;
Kang, C.; Kim, J.S. Gemcitabine-coumarin-biotin conjugates: a
target specific theranostic anticancer prodrug. J. Am. Chem. Soc.,
2013, 135(11), 4567-4572.
http://dx.doi.org/10.1021/ja401350x PMID: 23461361
[57] Khaghanzadeh, N.; Samiei, A.; Ramezani, M.; Mojtahedi, Z.;
Hosseinzadeh, M.; Ghaderi, A. Umbelliprenin induced production
of IFN-γ and TNF-α, and reduced IL-10, IL-4, Foxp3 and TGF-β in
a mouse model of lung cancer. Immunopharmacol. Immunotoxicol.,
2014, 36(1), 25-32.
http://dx.doi.org/10.3109/08923973.2013.863912 PMID: 24325354
[58] Karthik, S.; Puvvada, N.; Kumar, B.N.; Rajput, S.; Pathak, A.;
Mandal, M.; Singh, N.D. Photoresponsive coumarin-tethered multi-
functional magnetic nanoparticles for release of anticancer drug.
ACS Appl. Mater. Interfaces, 2013, 5(11), 5232-5238.
http://dx.doi.org/10.1021/am401059k PMID: 23730930
[59] Dlugosz, A.; Gach-Janczak, K.; Szymanski, J.; Deredas, D.; Kraw-
czyk, H.; Janecki, T.; Janecka, A. Anticancer properties of a new
hybrid analog AD-013 combining a coumarin scaffold with an α-
methylene-δ-lactone motif. Anti-Cancer Agents Med. Chem., 2018,
18(3), 450-457.
[60] Lalitha, K.; Nagarajan, S. Strongly fluorescent organogels and self-
assembled nanostructures from pyrene coupled coumarin deriva-
tives: application in cell imaging. J. Mater. Chem. B Mater. Biol.
Med., 2015, 3(28), 5690-5701.
http://dx.doi.org/10.1039/C5TB00694E
[61] Ji, W.; Liu, G.; Wang, F.; Zhu, Z.; Feng, C. Galactose-decorated
light-responsive hydrogelator precursors for selectively killing can-
cer cells. Chem. Commun. (Camb.), 2016, 52(85), 12574-12577.
http://dx.doi.org/10.1039/C6CC05707A PMID: 27477036
[62] Gao, X.; Wang, S.; Wang, B.; Deng, S.; Liu, X.; Zhang, X.; Luo,
L.; Fan, R.; Xiang, M.; You, C.; Wei, Y.; Qian, Z.; Guo, G. Im-
proving the anti-ovarian cancer activity of docetaxel with biode-
gradable self-assembly micelles through various evaluations. Bio-
materials, 2015, 53, 646-658.
http://dx.doi.org/10.1016/j.biomaterials.2015.02.108 PMID:
25890760
[63] Xu, G.; Shi, C.; Guo, D.; Wang, L.; Ling, Y.; Han, X.; Luo, J.
Functional-segregated coumarin-containing telodendrimer nanocar-
riers for efficient delivery of SN-38 for colon cancer treatment.
Acta Biomater., 2015, 21, 85-98.
http://dx.doi.org/10.1016/j.actbio.2015.04.021 PMID: 25910639
[64] Paolini, M.; Poul, L.; Darmon, A.; Germain, M.; Pottier, A.; Levy,
L.; Vibert, E. A new opportunity for nanomedicines: Micellar cyto-
chrome P450 inhibitors to improve drug efficacy in a cancer ther-
apy model. Nanomedicine (Lond.), 2017, 13(5), 1715-1723.
http://dx.doi.org/10.1016/j.nano.2017.03.006 PMID: 28343019
[65] Aas, Z.; Babaei, E.; Hosseinpour Feizi, M.A.; Dehghan, G. Anti-
proliferative and apoptotic effects of dendrosomal farnesiferol C on
gastric cancer cells. Asian Pac. J. Cancer Prev., 2015, 16(13),
5325-5329. http://dx.doi.org/10.7314/APJCP.2015.16.13.5325
PMID: 26225673
[66] Bhattacharyya, S.S.; Paul, S.; De, A.; Das, D.; Samadder, A.; Bou-
jedaini, N.; Khuda-Bukhsh, A.R. Poly (lactide-co-glycolide) acid
nanoencapsulation of a synthetic coumarin: Cytotoxicity and bio-
distribution in mice, in cancer cell line and interaction with calf
thymus DNA as target. Toxicol. Appl. Pharmacol., 2011, 253(3),
270-281.
http://dx.doi.org/10.1016/j.taap.2011.04.010 PMID: 21549736
[67] Gangopadhyay, M.; Singh, T.; Behara, K.K.; Karwa, S.; Ghosh,
S.K.; Singh, N.D. Coumarin-containing-star-shaped 4-arm-
polyethylene glycol: targeted fluorescent organic nanoparticles for
dual treatment of photodynamic therapy and chemotherapy. Photo-
chem. Photobiol. Sci., 2015, 14(7), 1329-1336.
http://dx.doi.org/10.1039/C5PP00057B PMID: 26066468
[68] He, G.; Yang, L.; Qian, X.; Li, J.; Yuan, Z.; Li, C. A coumarin-
based fluorescence resonance energy transfer probe targeting ma-
trix metalloproteinase-2 for the detection of cervical cancer. Int. J.
Mol. Med., 2017, 39(6), 1571-1579.
http://dx.doi.org/10.3892/ijmm.2017.2974 PMID: 28487974
[69] Stefanello, T.F.; Couturaud, B.; Szarpak-Jankowska, A.; Fournier,
D.; Louage, B.; Garcia, F.P.; Nakamura, C.V.; De Geest, B.G.;
Woisel, P.; van der Sanden, B.; Auzély-Velty, R. Coumarin-
containing thermoresponsive hyaluronic acid-based nanogels as de-
livery systems for anticancer chemotherapy. Nanoscale, 2017,
9(33), 12150-12162.
http://dx.doi.org/10.1039/C7NR03964F PMID: 28805867
[70] Chaudhuri, A.; Venkatesh, Y.; Das, J.; Behara, K.K.; Mandal, S.;
Maiti, T.K.; Singh, N.P. Squaric acid-coumarin-chlorambucil: Pho-
toresponsive single-component fluorescent organic nanoconjugates
for self-monitored therapeutics. ACS Appl. Nano Mater., 2018,
1(11), 6312-6319.
http://dx.doi.org/10.1021/acsanm.8b01533
[71] Khorramizadeh, M.; Esmail-Nazari, Z.; Zarei-Ghaane, Z.; Shaki-
baie, M.; Mollazadeh-Moghaddam, K.; Iranshahi, M.; Shahverdi,
A. Umbelliprenin-coated Fe3O4 magnetite nanoparticles: Antipro-
liferation evaluation on human Fibrosarcoma cell line (HT-1080).
Mater. Sci. Eng. C, 2010, 30(7), 1038-1042.
http://dx.doi.org/10.1016/j.msec.2010.05.005
[72] Liu, P.Y.; Chang, D.C.; Lo, Y.S.; Hsi, Y.T.; Lin, C.C.; Chuang,
Y.C.; Lin, S.H.; Hsieh, M.J.; Chen, M.K. Osthole induces human
nasopharyngeal cancer cells apoptosis through Fas-Fas ligand and
mitochondrial pathway. Environ. Toxicol., 2018, 33(4), 446-453.
http://dx.doi.org/10.1002/tox.22530 PMID: 29319219
[73] Xu, X-M.; Zhang, Y.; Qu, D.; Feng, X-W.; Chen, Y.; Zhao, L.
Osthole suppresses migration and invasion of A549 human lung
cancer cells through inhibition of matrix metalloproteinase-2 and
Nanoformulations of Coumarins and the Hybrid Molecules of Co umarins Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 1815
matrix metallopeptidase-9 in vitro. Mol. Med. Rep., 2012, 6(5),
1018-1022.
http://dx.doi.org/10.3892/mmr.2012.1044 PMID: 22923177
[74] Le Zou, T.; Wang, H.F.; Ren, T.; Shao, Z.Y.; Yuan, R.Y.; Gao, Y.;
Zhang, Y.J.; Wang, X.A.; Liu, Y.B. Osthole inhibits the progres-
sion of human gallbladder cancer cells through JAK/STAT3 signal
pathway both in vitro and in vivo. Anticancer Drugs, 2019, 30(10),
1022-1030.
http://dx.doi.org/10.1097/CAD.0000000000000812 PMID:
31283543
[75] Ju, A.-H.; Gong, W.-J.; Su, Y.; Mou, Z.-B. Imperatorin shows
selective antitumor effects in SGC-7901 human gastric adenocarci-
noma cells by inducing apoptosis, cell cycle arrest and targeting
PI3K/Akt/m-TOR signalling pathway. J. BU ON.: Offic. J. Balkan
Union Oncol., 2017, 22(6), 1471-1476.
[76] Mi, C.; Ma, J.; Wang, K.S.; Zuo, H.X.; Wang, Z.; Li, M.Y.; Piao,
L.X.; Xu, G.H.; Li, X.; Quan, Z.S.; Jin, X. Imperatorin suppresses
proliferation and angiogenesis of human colon cancer cell by tar-
geting HIF-via the mTOR/p70S6K/4E-BP1 and MAPK path-
ways. J. Ethnopharmacol., 2017, 203, 27-38.
http://dx.doi.org/10.1016/j.jep.2017.03.033 PMID: 28341244
[77] Kawaii, S.; Tomono, Y.; Ogawa, K.; Sugiura, M.; Yano, M.; Yo-
shizawa, Y.; Ito, C.; Furukawa, H. Antiproliferative effect of
isopentenylated coumarins on several cancer cell lines. Anticancer
Res., 2001, 21(3B), 1905-1911.
PMID: 11497276
[78] Żamojć, K.; Zdrowowicz, M.; Hać, A.; Witwicki, M.; Rudnicki-
Velasquez, P.B.; Wyrzykowski, D.; Wiczk, W.; Chmurzyński, L.
Dihydroxy-substituted coumarins as fluorescent probes for nano-
molar-level detection of the 4-amino-TEMPO spin label. Int. J.
Mol. Sci., 2019, 20(15), 3802.
http://dx.doi.org/10.3390/ijms20153802 PMID: 31382639
[79] Gong, J.; Zhang, W.-G.; Feng, X.-F.; Shao, M.-J.; Xing, C. Aes-
culetin (6, 7-dihydroxycoumarin) exhibits potent and se-lective an-
titumor activity in human acute myeloid leukemia cells (THP-1)
via induction of mitochondrial mediated apop-tosis and cancer cell
migration inhibition. J. BU ON.: Offic. J. Balkan Union Oncol.,
2017, 22(6), 1563-1569.
[80] Arora, R.; Sawney, S.; Saini, V.; Steffi, C.; Tiwari, M.; Saluja, D.
Esculetin induces antiproliferative and apoptotic response in pan-
creatic cancer cells by directly binding to KEAP1. Mol. Cancer,
2016, 15(1), 64.
http://dx.doi.org/10.1186/s12943-016-0550-2 PMID: 27756327
[81] Tian, Q.; Wang, L.; Sun, X.; Zeng, F.; Pan, Q.; Xue, M. Scopoletin
exerts anticancer effects on human cervical cancer cell lines by
triggering apoptosis, cell cycle arrest, inhibition of cell invasion
and PI3K/AKT signalling pathway. J. BU ON.: Offic. J. Balkan
Union Oncol., 2019, 24(3), 997-1002.
[82] Tabana, Y.M.; Hassan, L.E.A.; Ahamed, M.B.K.; Dahham, S.S.;
Iqbal, M.A.; Saeed, M.A.; Khan, M.S.S.; Sandai, D.; Majid,
A.S.A.; Oon, C.E.; Majid, A.M. Scopoletin, an active principle of
tree tobacco (Nicotiana glauca) inhibits human tumor vasculariza-
tion in xenograft models and modulates ERK1, VEGF-A, and FGF-
2 in computer model. Microvasc. Res., 2016, 107, 17-33.
http://dx.doi.org/10.1016/j.mvr.2016.04.009 PMID: 27133199
[83] Rahman, M.A.; Kim, N-H.; Yang, H.; Huh, S-O. Angelicin induces
apoptosis through intrinsic caspase-dependent pathway in human
SH-SY5Y neuroblastoma cells. Mol. Cell. Biochem., 2012, 369(1-
2), 95-104.
http://dx.doi.org/10.1007/s11010-012-1372-1 PMID: 22766766
[84] Wu, H-J.; Wu, H-B.; Zhao, Y-Q.; Chen, L-J.; Zou, H-Z. Bergamot-
tin isolated from Citrus bergamia exerts in vitro and in vivo anti-
tumor activity in lung adenocarcinoma through the induction of
apoptosis, cell cycle arrest, mitochondrial membrane potential loss
and inhibition of cell migration and invasion. Oncol. Rep., 2016,
36(1), 324-332.
http://dx.doi.org/10.3892/or.2016.4833 PMID: 27222242
[85] Gismondi, A.; Nanni, V.; Reina, G.; Orlanducci, S.; Terranova,
M.L.; Canini, A. Nanodiamonds coupled with 5,7-
dimethoxycoumarin, a plant bioactive metabolite, interfere with the
mitotic process in B16F10 cells altering the actin organization. Int.
J. Nanomedicine, 2016, 11, 557-574.
http://dx.doi.org/10.2147/IJN.S96614 PMID: 26893562
[86] Wu, J-Y.; Li, Y-J.; Liu, T-T.; Ou, G.; Hu, X-B.; Tang, T-T.; Wang,
J-M.; Liu, X-Y.; Xiang, D-X. Microemulsions vs chitosan deriva-
tive-coated microemulsions for dermal delivery of 8-
methoxypsoralen. Int. J. Nanomedicine, 2019, 14, 2327-2340.
http://dx.doi.org/10.2147/IJN.S191940 PMID: 31015760
[87] La, V.D.; Zhao, L.; Epifano, F.; Genovese, S.; Grenier, D. Anti-
inflammatory and wound healing potential of citrus auraptene. J.
Med. Food, 2013, 16(10), 961-964.
http://dx.doi.org/10.1089/jmf.2013.0029 PMID: 24070132
[88] Yang, X.W.; Xu, B.; Ran, F.X.; Wang, R.Q.; Wu, J.; Cui, J.R.
Inhibitory effects of 11 coumarin compounds against growth of
human bladder carcinoma cell line E-J in vitro. J. Chin. Integr.
Med., 2007, 5(1), 56-60.
http://dx.doi.org/10.3736/jcim20070111 PMID: 17214937
[89] Dong, L.; Xu, W-W.; Li, H.; Bi, K-H. In vitro and in vivo antican-
cer effects of marmesin in U937 human leukemia cells are medi-
ated via mitochondrial-mediated apoptosis, cell cycle arrest, and
inhibition of cancer cell migration. Oncol. Rep., 2018, 39(2), 597-
602.
PMID: 29251335
[90] Suhaimi, S.A.; Hong, S.L.; Abdul Malek, S.N. Rutamarin, an ac-
tive constituent from Ruta angustifolia Pers., induced apoptotic cell
death in the HT29 colon adenocarcinoma cell line. Pharmacogn.
Mag., 2017, 13(Suppl. 2), S179-S188.
http://dx.doi.org/10.4103/pm.pm_432_16 PMID: 28808378
[91] Fakai, M.I.; Karsani, S.A.; Malek, S.N.A. Chalepin and rutamarin
isolated from Ruta angustifolia inhibit cell growth in selected can-
cer cell lines (MCF7, MDA-MB-231, HT29, AND HCT116). J. In-
form., 2017, 2(5), 8-17.
[92] Fakai, M.I.; Abd Malek, S.N.; Karsani, S.A. Induction of apoptosis
by chalepin through phosphatidylserine externalisations and DNA
fragmentation in breast cancer cells (MCF7). Life Sci., 2019, 220,
186-193.
http://dx.doi.org/10.1016/j.lfs.2019.01.029 PMID: 30682342
[93] Richardson, J.S.M.; Sethi, G.; Lee, G.S.; Malek, S.N.A. Chalepin:
Isolated from Ruta angustifolia L. Pers induces mitochondrial me-
diated apoptosis in lung carcinoma cells. BMC Complement. Al-
tern. Med., 2016, 16(1), 389.
http://dx.doi.org/10.1186/s12906-016-1368-6 PMID: 27729078
[94] Richardson, J.S.M.; Aminudin, N.; Abd Malek, S.N. Chalepin: A
compound from Ruta angustifolia L. pers exhibits cell cycle arrest
at S phase, suppresses nuclear factor-kappa B (NF-κB) pathway,
signal transducer and activation of transcription 3 (STAT3) phos-
phorylation and extrinsic apoptotic pathway in non-small cell lung
cancer carcinoma (A549). Pharmacogn. Mag., 2017, 13(Suppl. 3),
S489-S498.
http://dx.doi.org/10.4103/pm.pm_13_17 PMID: 29142404
[95] Kang, J.I.; Hong, J-Y.; Choi, J.S.; Lee, S.K. Columbianadin inhib-
its cell proliferation by inducing apoptosis and necroptosis in
HCT116 colon cancer cells. Biomol. Ther. (Seoul), 2016, 24(3),
320-327.
http://dx.doi.org/10.4062/biomolther.2015.145 PMID: 27098859
[96] Lei, P.; Liao, C.; Chen, J.; Zhou, M. In vitro and in vivo growth
inhibition of human leukemia cells by Nodakenetin are mediated
via mitochondrial apoptosis, cell cycle arrest and inhibition of cell
migration and invasion. J. BU ON.: Offic. J. Balkan Union Oncol.,
2019, 24(3), 1219-1224.
[97] Znati, M.; Debbabi, M.; Romdhane, A.; Ben Jannet, H.; Bouajila, J.
Synthesis of new anticancer and anti-inflammatory isoxazolines
and aziridines from the natural (-)-deltoin. J. Pharm. Pharmacol.,
2018, 70(12), 1700-1712.
http://dx.doi.org/10.1111/jphp.13013 PMID: 30229910
[98] Ben Salem, S.; Jabrane, A.; Harzallah-Skhiri, F.; Ben Jannet, H.
New bioactive dihydrofuranocoumarins from the roots of the Tuni-
sian Ferula lutea (Poir.) Maire. Bioorg. Med. Chem. Lett., 2013,
23(14), 4248-4252.
http://dx.doi.org/10.1016/j.bmcl.2013.04.081 PMID: 23746477
[99] He, F.; Wang, M.; Gao, M.; Zhao, M.; Bai, Y.; Zhao, C. Chemical
composition and biological activities of Gerbera anandria. Mole-
cules, 2014, 19(4), 4046-4057.
http://dx.doi.org/10.3390/molecules19044046 PMID: 24699147
[100] Lü, J.; Zhang, J.; Li, L.; Jiang, C.; Xing, C. Cancer chemopreven-
tion with Korean Angelica: Active compounds, pharmacokinetics,
and human translational considerations. Curr. Pharmacol. Rep.,
2015, 1(6), 373-381.
http://dx.doi.org/10.1007/s40495-015-0033-y PMID: 26623248
[101] Zhang, J.; Li, L.; Jiang, C.; Xing, C.; Kim, S.-H.; Lu, J. Anti-cancer
and other bioactivities of Korean Angelica gigas Nakai (AGN) and
1816 Anti-Cancer Agents in Medicinal Chemistry, 2020, Vol. 20, No. 15 Yerer et al.
its major pyranocoumarin compounds. Anti-Cancer Agents Med.
Chem., 2012, 12(10), 1239-1254.
[102] Reddy, C.S.; Kim, S.C.; Hur, M.; Kim, Y.B.; Park, C.G.; Lee,
W.M.; Jang, J.K.; Koo, S.C. Natural Korean medicine dang-gui:
Biosynthesis, effective extraction and formulations of major active
pyranocoumarins, their molecular action mechanism in cancer, and
other biological activities. Molecules, 2017, 22(12), 2170.
http://dx.doi.org/10.3390/molecules22122170 PMID: 29215592
[103] Tang, S-N.; Zhang, J.; Wu, W.; Jiang, P.; Puppala, M.; Zhang, Y.;
Xing, C.; Kim, S-H.; Jiang, C.; Lü, J. Chemopreventive effects of
korean angelica versus its major pyranocoumarins on two lineages
of transgenic adenocarcinoma of mouse prostate carcinogenesis.
Cancer Prev. Res. (Phila.), 2015, 8(9), 835-844.
http://dx.doi.org/10.1158/1940-6207.CAPR-15-0051 PMID:
26116406
[104] Rasul, A.; Khan, M.; Yu, B.; Ma, T.; Yang, H. Xanthoxyletin, a
coumarin induces S phase arrest and apoptosis in human gastric
adenocarcinoma SGC-7901 cells. Asian Pac. J. Cancer Prev.,
2011, 12(5), 1219-1223.
PMID: 21875271
[105] Lee, J.; Lee, Y.J.; Kim, J.; Bang, O-S. Pyranocoumarins from root
extracts of Peucedanum praeruptorum Dunn with multidrug resis-
tance reversal and anti-inflammatory activities. Molecules, 2015,
20(12), 20967-20978.
http://dx.doi.org/10.3390/molecules201219738 PMID: 26610461
[106] Wu, M-H.; Lin, C-L.; Chiou, H-L.; Yang, S-F.; Lin, C-Y.; Liu, C-
J.; Hsieh, Y-H. Praeruptorin A inhibits human cervical cancer cell
growth and invasion by suppressing MMP-2 expression and
ERK1/2 signaling. Int. J. Mol. Sci., 2017, 19(1), 10.
http://dx.doi.org/10.3390/ijms19010010 PMID: 29267213
[107] Liang, T.; Yue, W.; Li, Q. Chemopreventive effects of Peuceda-
num praeruptorum DUNN and its major constituents on SGC7901
gastric cancer cells. Molecules, 2010, 15(11), 8060-8071.
http://dx.doi.org/10.3390/molecules15118060 PMID: 21063269
[108] Kim, J.; Kim, H-Y.; Hong, S.; Shin, S.; Kim, Y.A.; Kim, N.S.;
Bang, O-S. A new herbal formula BP10A exerted an antitumor ef-
fect and enhanced anticancer effect of irinotecan and oxaliplatin in
the colon cancer PDTX model. Biomed. Pharmacother., 2019, 116,
108987
http://dx.doi.org/10.1016/j.biopha.2019.108987 PMID: 31112870
[109] Jun, N.J.; Kim, S.-C.; Song, E.-Y.; Jang, K.C.; Lee, D.S.; Cho, S.K.
Isolation of anticancer compounds from Peucedanum japonicum
Thunb. Roots. 2014, 27(3), 215-222.
[110] Jung, S.; Moon, H-I.; Lee, B.S.; Kim, S.; Quynh, N.T.N.; Yu, J.;
Le, D.T.; Sandag, Z.; Lee, H.; Lee, H.; Anh, N.H.; Yang, Y.; Lim,
J.S.; Kim, K.I.; Lee, M.S. Anti-cancerous effect of cis-khellactone
from Angelica amurensis through the induction of three pro-
grammed cell deaths. Oncotarget, 2018, 9(24), 16744-16757.
http://dx.doi.org/10.18632/oncotarget.24686 PMID: 29682182
[111] Arbab, I.A.; Looi, C.Y.; Abdul, A.B.; Cheah, F.K.; Wong, W.F.;
Sukari, M.A.; Abdullah, R.; Mohan, S.; Syam, S.; Arya, A. Den-
tatin induces apoptosis in prostate cancer cells via Bcl-2, Bcl-xL,
Survivin downregulation, caspase-9,-3/7 activation, and NF-κB in-
hibition. Evid.-Based Complement. Altern. Med., 2012, 2012, Arti-
cle ID 856029.
[112] Sharif, N.M.; Mustahil, N.; Noor, N .M.; Sukari, M.; Rahmani, M.;
Taufiq-Yap, Y.; Ee, G. Cytotoxic constituents of Clausena exca-
vata. Afr. J. Biotechnol., 2011, 10(72), 16337-16341.
[113] Yao, G-D.; Cheng, Z-Y.; Shang, X-Y.; Gao, P-Y.; Huang, X-X.;
Song, S-J. Coumarins from the bark of Juglans mandshurica exhib-
ited anti-hepatoma activities via inducing apoptosis. J. Asian Nat.
Prod. Res., 2017, 19(11), 1134-1142.
http://dx.doi.org/10.1080/10286020.2017.1292256 PMID:
28276763
[114] Chun, J.; Kim, J.; Kim, Y.S. 3′, 4′-Disenecioylkhellactone from
Peucedanum japonicum Thunb. induces apoptosis mediated by in-
hibiting STAT3 signaling in human gastric cancer cells. Korean J.
Pharmacogn., 2018, 49(3), 225-230.
[115] Maleki, D.; Kyoomehr, P.; Rajabi, A.; Amin, G.; Azizi, E. Cyto-
toxic activity of Ammi visnaga (L.) Lam. against T47D (breast duc-
tal carcinoma) cell line. North Khorasan Univ. Med. Sci., 2012.
http://journals.nkums.ac.ir/index.php/ndnkh/article/viewFile/292/47
2. Accessed February 27, 2017.
[116] Shen, X.; Chen, G.; Zhu, G.; Cai, J.; Wang, L.; Hu, Y.; Fong, W.F.
3′-O, 4′-O-aromatic acyl substituted 7,8-pyranocoumarins: a new
class of P-glycoprotein modulators. J. Pharm. Pharmacol., 2012,
64(1), 90-100.
http://dx.doi.org/10.1111/j.2042-7158.2011.01378.x PMID:
22150676
[117] Kathuria, A.; Jalal, S.; Tiwari, R.; Shirazi, A.N .; Gupta, S.; Kumar,
S.; Parang, K.; Sharma, S.K. Substituted coumarin derivatives:
Synthesis and evaluation of antiproliferative and Src kinase inhibi-
tory activities. Chem. Biol. Interface, 2011, 1, 279-296.
[118] Ren, L.; Du, X.; Hu, M.; Yan, C.; Liang, T.; Li, Q. Design, synthe-
sis and antitumor activity of novel 4-methyl-(3‘S,4’S)-cis-
khellactone derivatives. Molecules, 2013, 18(4), 4158-4169.
http://dx.doi.org/10.3390/molecules18044158 PMID: 23567363
[119] Jóźwiak, M.; Struga, M.; Roszkowski, P.; Filipek, A.; Nowicka, G.;
Olejarz, W. Anticancer effects of alloxanthoxyletin and fatty acids
esters - In vitro study on cancer HTB-140 and A549 cells. Biomed.
Pharmacother., 2019, 110, 618-630.
http://dx.doi.org/10.1016/j.biopha.2018.12.005 PMID: 30544062
[120] Ostrowska, K.; Olejarz, W.; Wrzosek, M.; Głuszko, A.; Nowicka,
G.; Szczepański, M.; Materek, I.B.; Kozioł, A.E.; Struga, M. Anti-
cancer effects of O-aminoalkyl derivatives of alloxanthoxyletin and
seselin. Biomed. Pharmacother., 2017, 95, 1412-1424.
http://dx.doi.org/10.1016/j.biopha.2017.09.050 PMID: 28946189
[121] Shi, W.; Zhang, J.; Bao, N.; Guan, F.; Chen, L.; Sun, J. Design,
synthesis, and cytotoxic evaluation of novel scopoletin derivatives.
Chem. Biol. Drug Des., 2018, 91(2), 641-646.
http://dx.doi.org/10.1111/cbdd.13120 PMID: 29052945
... It is a naturally occurring flavonoid compound found in honey, propolis, passion flowers, Passiflora caerulea and Passiflora incarnata. Flavonoids are plant-based poly-phenolic phytochemicals that are reported to have anti-cancer and chemo-preventive properties [7]. Phytochemicals such as coumarin, gallic acid's anticancer activities are unaffected by a number of biological processes, including the activation of programmed cell death, cell cycle arrest, reluctance of tumour migration, and inflammation and now CH is being extensively researched for its anticancer effects and till now a plethora of evidence-based studies are predicting a mixed overview on its anticancer potential [8,9]. ...
Article
In recent times, there have been notable advancements in comprehending the potential anti-cancer effects of chrysin (CH), a naturally occurring flavonoid compound found abundantly in various plant sources like honey, propolis, and certain fruits and vegetables. This active compound has garnered significant attention due to its promising therapeutic qualities and minimal toxicity. CH's ability to combat cancer arises from its multifaceted mechanisms of action, including the initiation of apoptosis and the inhibition of proliferation, angiogenesis, metastasis, and cell cycle progression. CH also displays potent antioxidant and anti-Explor Target Antitumor Ther. 2024;5:477-94 | https://doi.org/10.37349/etat.2024.00230 Page 478 inflammatory properties, effectively counteracting the harmful molecules that contribute to DNA damage and the development of cancer. Furthermore, CH has exhibited the potential to sensitize cancer cells to traditional chemotherapy and radiotherapy, amplifying the effectiveness of these treatments while reducing their negative impact on healthy cells. Hence, in this current review, the composition, chemistry, mechanisms of action, safety concerns of CH, along with the feasibility of its nanoformulations. To conclude, the recent investigations into CH's anti-cancer effects present a compelling glimpse into the potential of this natural compound as a complementary therapeutic element in the array of anti-cancer approaches, providing a safer and more comprehensive method of combating this devastating ailment.
... Sharma et al. revealed that coumarin caused a high percentage of egg and larval mortality of the potato tuber moth, Phthorimaea operculella Zell [35]. Moreover, a total of four coumarins isolated from the crude seed extract of Mammea siamensis-surangin B, surangin C, mammea E/BB, and mammea E/BC were reported to have insecticidal properties [36]. These results indicated that coumarins are highly phytotoxic and effective in controlling pests. ...
Article
Full-text available
Coumarin and its derivatives are plant-derived compounds that exhibit potent insecticidal properties. In this study, we found that natural coumarin significantly inhibited the growth and development of Spodoptera litura larvae through toxicological assay. By transcriptomic sequencing, 80 and 45 differentially expressed genes (DEGs) related to detoxification were identified from 0 to 24 h and 24 to 48 h in S. litura after coumarin treatment, respectively. Enzyme activity analysis showed that CYP450 and acetylcholinesterase (AChE) activities significantly decreased at 48 h after coumarin treatment, while glutathione S-transferases (GST) activity increased at 24 h. Silencing of SlCYP324A16 gene by RNA interference significantly increased S. litura larval mortality and decreased individual weight after treatment with coumarin. Additionally, the expression levels of DEGs involved in glycolysis and tricarboxylic acid (TCA) cycle were inhibited at 24 h after coumarin treatment, while their expression levels were upregulated at 48 h. Furthermore, metabonomics analysis identified 391 differential metabolites involved in purine metabolism, amino acid metabolism, and TCA cycle from 0 to 24 h after treated with coumarin and 352 differential metabolites associated with ATP-binding cassette (ABC) transporters and amino acid metabolism. These results provide an in-depth understanding of the toxicological mechanism of coumarin on S. litura.
Article
Full-text available
Herein, a rapid, clean, less expensive and environmentally friendly route to a novel series of coumarins bearing thiazoles or 1,3,4-thiadiazoles was developed via grinding method under solvent conditions. Thus, 6-bromo-3-(2-bromoacetyl)-2-chromen-2-one was treated with cyanothioacetamide to produce thiazole-2-acetonitrile derivative 2, which was then transformed to iminocoumarins by reacting with hydroxyaldehydes. Hydrolysis of iminocoumarins by conc. HCl furnished coumarins. Furthermore, treatment of compound 2 with phenylisothiocyanate produced thioanilide which interacts with hydrazonoyl chloride and/or α-halocarbonyl compounds afforded 1,3,4-thiadiazole and thiophene derivatives, consequently. All the newly prepared coumarins were screened against six pathogenic microorganisms. The results indicated that compounds 7 and 12 were the most effective against Bacillus pumilis, while compound 4b was highly active against Streptococcus faecalis. Also, compound 5b was highly active against Enterobacter cloacae. Compounds 2 and 7 were higher active compared with reference drug ketoconazole against fungi Saccharomyces cerevisiae.
Article
Full-text available
Globally, lung cancer is a significant public health concern due to its role as the leading cause of cancer-related mortalities. The promising target of EGFR for lung cancer treatment has been identified, providing a potential avenue for more effective therapies. The purpose of the study was to design a library of 1843 coumarin-1,2,3-triazole hybrids and screen them based on a designed pharmacophore to identify potential inhibitors targeting EGFR in lung cancer with minimum or no side effects. Pharmacophore-based screening was carried out and 60 hits were obtained. To gain a better understanding of the binding interactions between the compounds and the targeted receptor, molecular docking was conducted on the 60 screened compounds. In-silico ADME and toxicity studies were also conducted to assess the drug-likeness and safety of the identified compounds. The results indicated that coumarin-1,2,3-triazole hybrids COUM-0849, COUM-0935, COUM-0414, COUM-1335, COUM-0276, and COUM-0484 exhibit dock score of − 10.2, − 10.2, − 10.1, − 10.1, − 10, − 10 while reference molecule − 7.9 kcal/mol for EGFR (PDB ID: 4HJO) respectively. The molecular docking and molecular dynamics simulations revealed that the identified compounds formed stable interactions with the active site of EGFR, indicating their potential as inhibitors. The in-silico ADME and toxicity studies showed that the compounds had favorable drug-likeness properties and low toxicity, further supporting their potential as therapeutic agents. Finally, we performed DFT studies on the best-selected ligands to gain further insights into their electronic properties. The findings of this study provide important insights into the potential of coumarin-1,2,3-triazole hybrids as promising EGFR inhibitors for the management of lung cancer. Graphical abstract
Article
Full-text available
Plant secondary metabolites, including furanocoumarins, have attracted attention for decades as active molecules with therapeutic potential, especially those occurring in a limited number of species as evolutionarily specific and chemotaxonomically important. The most famous methoxyfuranocoumarins (MFCs), bergapten, xanthotoxin, isopimpinellin, phellopterin, byakangeli-col, byakangelicin, isobergapten, pimpinellin, sphondin, as well as rare ones such as peucedanin and 8-methoxypeucedanin, apaensin, cnidilin, moellendorffiline and dahuribiethrins, have recently been investigated for their various biological activities. The α-glucosidase inhibitory activity and antioxidant potential of moellendorffiline, the antiproliferative and proapoptotic properties of non-UV-activated bergapten and xanthotoxin, the effect of MFC on the activity of tyrosinase, acetyl-and butylcholinesterase, and the role of these compounds as adjuvants in anticancer and antibacterial tests have been confirmed. The anticonvulsant effects of halfordin, the antidepressant effects of xanthotoxin, and the antiadipogenic, neuroprotective, anti-amyloid-β, and anti-inflammatory (via increasing SIRT 1 protein expression) properties of phellopterin, as well as the activity of sphondin against hepatitis B virus, have also attracted interest. It is worth paying attention to the agonistic effect of xanthotoxin on bitter taste receptors (TAS2Rs) on cardiomyocytes, which may be important in the future treatment of tachycardia, as well as the significant anti-inflammatory activity of dahuribiethrins. It should be emphasized that MFCs, although in many cases isolated for the first time many years ago, are still of great interest as bioactive molecules. The aim of this review is to highlight key recent developments in the study of the diverse biological activities of MFCs and attempt to highlight promising directions for their further research. Where possible, descriptions of the mechanisms of action of MFC are provided, which is related to the constantly discovered therapeutic potential of these molecules. The review covers the results of experiments from the last ten years (2014-2023) conducted on isolated natural cMFCs and includes the activity of molecules that have not been activated by UV rays.
Article
Full-text available
Purpose: Cervical cancer causes considerable mortality in women world over and the current treatment options create severe adverse effects. Hence, there is an urgent need to develop novel and efficient treatment regimens for cervical cancer. Herein, we examined the anticancer effects of a natural coumarin, Scopoletin, against a panel of cervical cancer cell lines. Methods: The antiproliferative effect of Scopoletin was examined by cell counting and colony formation assays. Apoptosis was detected by acridine orange (AO) ethidium promide (EB) staining. Cell cycle distribution was determined by flow cytometry. Cell invasion was examined by Boyden chamber assay. Protein expression was checked by western blotting. Results: Scopoletin inhibited the growth of all the cell lines and the IC50 ranged between 7.5 to 25 µM. Nonetheless, the cytotoxic effects of Scopoletin were comparatively negligible against the normal cells with an IC50 of 90 µM. Investigation of the mechanism of action, revealed that the anticancer effects of Scopoletin against the HeLa cervical cancer cells were due to induction of apoptotic cell death as indicated AO/EB staining. Scopoletin treatment also resulted in enhancement of the Bax, Caspase 3, 8 and 9 expression and decline of the Bcl-2 expression. Scopoletin also blocked the HeLa cells at G2/M checkpoint of the cell cycle. Furthermore, cell invasion assay revealed that Scopoletin inhibited the migration of the HeLa cells concentration-dependently. PI3K/AKT is an imperative pathway involved in theproliferation and tumorigenesis of cancer cells and herein it was found that Scopoletin could inhibit this pathway. Conclusion: Taken together, Scopoletin may prove essential in the development of novel treatment regimes for cervical cancer.
Article
Full-text available
Purpose: To study the anticancer potential of a plant-derived coumarin, Nodakenetin, against acute lymphocytic leukemia HL-60 cells. Methods: The proliferation rate of the leukemia cells was checked by CCK-8 assay. Apoptotic cell death was studied by acridine orange (AO)/ethidium bromide (EB) staining. Cell cycle analysis was performed by flow cytometery. Cell migration and invasion were checked by transwell assay. Protein expression were determined by immuno blotting. Xenografted mice models were used for in vivo evaluation of Nodakenetin. Results: Nodakenetin could significantly inhibit the proliferation of the all the leukemia cells. The anticancer activity of Nodakenetin against the HL-60 cells was found to be due to G2/M cell cycle arrest and induction of apoptosis. Nodakenetin prompted mitochondrial apoptosis which was also associated with alteration in the apoptosis-related protein expression (Bax and Bcl-2). It was also observed that Nodakenetin could inhibit the migration and invasion of the leukemia cells in a concentration-dependent manner. The effects of the Nodakenetin were also investigated in vivo in xenografted mice models and it was found that this molecule could inhibit the growth of xenografted tumors. Conclusions: These results indicate Nodakenetin significantly inhibits the growth of leukemia cells in vitro and in vivo and may be a valuable molecule in the management of leukemia, and as such needs further in depth investigations.
Article
Full-text available
This paper reports on dihydroxycoumarins as fluorescent probes suitable for the detection and determination of the nitroxide radical, namely 4-amino-TEMPO. Since 4-amino-TEMPO is used as a spin label for the detection of various radicals and damage caused by these species, its determination under physiological conditions might help us to understand the mechanism of the oxidative stress. Among different coumarins studied, only dihydroxy-substituted derivatives show high sensitivity, specificity, and selectivity for the nitroxide radical. In this assay, dihydroxy-substituted coumarins under the action of 4-amino-TEMPO show a very fast and significant increase in fluorescence intensity and lifetime. Among them 6,7-dihydroxycoumarin (esculetin) exhibits the strongest fluorescence enhancement (up to 40 times), with an estimated limit of detection equal to 16.7 nM—a significantly lower value when compared with UV-Vis or electron paramagnetic resonance (EPR) spectroscopy. The method is characterized by an easy procedure of sample preparation and very short time of analysis. The mechanism of the interaction between 6,7-dihydroxycoumarin and 4-amino-TEMPO has been examined with the use of a series of complementary techniques, such as steady-state and time-resolved fluorescence spectroscopy, UV-Vis spectroscopy, electron paramagnetic resonance spectroscopy, potentiometric titration, and high-performance liquid chromatography. It has been proven that the only route of the reaction in the system studied is a proton transfer from the molecule of esculetin to the amino group of the nitroxide. Biological studies performed on prostate cancer cells, breast cancer cells, and normal skin fibroblasts revealed significant anticancer properties of 6,7-dihydroxycoumarin, which caused a considerable decrease in the viability and number of cancer cells, and affected their morphology, contrary to normal fibroblasts. Furthermore, the experiment performed on prostate cancer cells showed that fluorescence emission of esculetin is closely related to intracellular pH—the higher pH, the higher observed fluorescence intensity (in accordance with a chemical experiment). On the other hand, the studies performed in different pH levels revealed that when pH of the solution increases, the observed fluorescence intensity enhancement under the action of 4-amino-TEMPO decreases (better sensing properties of esculetin towards the nitroxide in lower pH).
Article
Full-text available
BP10A is a novel two-herb medicine formula, consisting of Descurainiae sophia Semen and Peucedani praeruptorum Radix. This study was done to evaluate the antitumor efficacy of BP10A and its effect on the efficacy of the anticancer drugs oxaliplatin and irinotecan (CPT-11) in a colon tumor xenograft model. Chemical constituents from the ethanol extracts of BP10A were characterized with the ultra-performance liquid chromatography (UPLC) and each constituent was quantified with the UPLC-diode array detector method. Our study showed that BP10A exerted the cytotoxic effects in two colorectal cancer cell lines and its combination treatments with oxaliplatin or CPT-11 remarkably increased the in vitro cytotoxicity of each cancer drug assessed by the Ez-cytox assay. The in vivo antitumor activity of BP10A was evaluated in three colon cancer patient-derived tumor xenograft (PDTX) models with different genetic backgrounds. Oral administration with BP10A (250 and 500 mg/kg, daily) delayed tumor growth by 34-70% in the all PDTX models. Similarly, intraperitoneal injection of oxaliplatin (6 mg/kg) or CPT-11 (20 mg/kg) also suppressed tumor growth by 31.8-60.5% or by 24.3-50.4%, respectively. Furthermore, the combination treatment of BP10A with oxaliplatin or CPT-11 remarkably enhanced the antitumor activity of each anti-cancer drug and delayed tumor growth by 47.1-74.6% or by 74.4-82.9%, respectively. In accordance with the antitumor activity, the Ki-67 expression for tumor cell proliferation and the CD31 for angiogenesis were decreased, and TUNEL staining for tumor cell apoptosis was remarkably increased by the co-treatment of BP10A and the anticancer drugs as well as by each treatment of BP10A, oxaliplatin or CPT-11. Conclusively, BP10A has a strong tumor inhibitory effect against colon cancer and a synergistic effect with anticancer drugs, suggesting that BP10A could be considered as a good therapeutic candidate for the treatment of colon cancer.
Article
Full-text available
Osthole is an antitumor compound, which effect on Gallbladder cancer (GBC) has been not elucidated. This study focused on its anti-GBC effect and mechanism both in vitro and in vivo. The antiproliferation effect on cell lines NOZ and SGC-996 were measured by cell counting kit-8 (CCK-8) and colony formation assay. The effects on cell apoptosis and cell cycle were investigated by flow cytometry assay. The migration effect was checked by transwell assay and the expressions of proteins were examined by Western Blots. Also, we did an in-vivo experiment by intraperitoneal injection of osthole in nude mice. The results showed that cell proliferation and viability were inhibited in a dose- and time-dependent manner. The similar phenomenon was also found in vivo. Flow cytometric assay confirmed that osthole inhibited cells proliferation via inducing apoptosis and G2/M arrest. Transwell assay indicated that osthole inhibited the migration in a dose-dependent manner. Expression of key proteins related with apoptosis and cell cycle were testified after osthole treatment. Also, we found the key proteins involved in the JAK/STAT3 signal way decreased after osthole treatment. This study suggested that osthole can inhibit the progression of human GBC cell lines, thus maybe a potential drug for GBC treatment.
Article
Full-text available
Keeping in view various pharmacological attributes of indole and coumarin derivatives, a new series of indolindione–coumarin molecular hybrids was rationally designed and synthesized. All synthesized hybrid molecules were evaluated for their antimicrobial potential against Gram-negative bacterial strains (Escherichia coli and Salmonella enterica), Gram-positive bacterial strains (Staphylococcus aureus and Mycobacterium smegmatis), and four fungal strains (Candida albicans, Alternaria mali, Penicillium sp., and Fusarium oxysporum) by using the agar gel diffusion method. Among all synthetics, compounds K-1 and K-2 were found to be the best antimicrobial agents with the minimum inhibitory concentration values of 30 and 312 μg/mL, against Penicillium sp. and S. aureus, respectively. The biological data revealed some interesting facts about the structure–activity relationship which state that the electronic environment on the indolinedione moiety and carbon chain length between indolinedione and triazole moieties considerably affect the antimicrobial potential of the synthesized hybrids. Various types of binding interactions of K-2 within the active site of S. aureus dihydrofolate reductase were also streamlined by molecular modeling studies, which revealed the possible mechanism for potent antibacterial activity of the compound.
Article
Full-text available
Molecular containers that provide both stimuli-responsive assembly/disassembly properties and wide-ranging host capabilities in aqueous medium still remain a current synthetic challenge. Herein we report polyaromatic nanocapsules assembled from V-shaped amphiphilic molecules bearing a photoresponsive ortho-dianthrylbenzene unit in water. Unlike previously reported supramolecular capsules and cages, the nanocapsules quickly and quantitatively disassemble into monomeric species by a non-invasive light stimulus through structural conversion from the open to the closed form of the amphiphiles. Regeneration of the nanocapsules is demonstrated by light irradiation or heating of the closed amphiphiles. With the aid of the wide-ranging host capability, the photo-induced release of various encapsulated guest molecules (e.g., Nile red, Cu(II)-phthalocyanine, and fullerene C60) can be achieved by using the present nanocapsule in water. This feature can furthermore be utilized to switch the fluorescence of encapsulated coumarin guests through their controlled release.
Article
Full-text available
Background: 8-methoxypsoralen (8-MOP) is one of the most commonly utilized drugs in psoralen-ultraviolet A therapy for treatment of vitiligo. However, poor skin retention and systemic side effects limit the clinical application of 8-MOP. Methods: Microemulsions (MEs) and chitosan derivative-coated 8-MOP MEs were developed and compared for dermal delivery of 8-MOP. Ex vivo skin retention/permeation study was performed to select the ME formulation with the highest retention:permeation ratio. Four different chitosan-coated MEs were prepared and compared with the ME formulation for their ability to distribute 8-MOP in the skin. Results: Among various ME formulations developed, a formulation containing 2.9% ethyl oleate, 17.2% Cromophor EL35, 8.6% ethanol and 71.3% water showed the highest ex vivo skin retention:permeation ratio (1.98). Of four chitosan-coated MEs prepared, carboxymethyl chitosan-coated MEs (CC-MEs) and hydroxypropyl chitosan-coated MEs (HC-MEs) showed higher ex vivo skin retention:permeation ratio (1.46 and 1.84). and were selected for in vivo pharmacokinetic study. AUCskin (0-12 h) for 8-MOP MEs (4578.56 h·ng·mL-1) was higher than HC-MEs (3422.47 h·ng·mL-1), CC-MEs (2808.51 h·ng·mL-1) and tincture (1500.16 h·ng·mL-1). Also, AUCplasma (0-12 h) for MEs (39.35±13.90 h·ng·mL-1) was significantly lower than HC-MEs (66.32 h·ng·mL-1), CC-MEs (59.70 h·ng·mL-1) and tincture (73.02 h·ng·mL-1). Conclusion: These combined results suggested that the MEs developed could be a promising and safe alternative for targeted skin delivery of 8-MOP.
Article
Full-text available
The growing interest in anticancer hybrids in the last few years has resulted in a great number of reports on hybrid design, synthesis and bioevaluation. Many novel multi-target-directed drug candidates were synthesized, and their biological activities were evaluated. For the design of anticancer hybrid compounds, the molecules of stilbenes, aromatic quinones, and heterocycles (benzimidazole, imidazole, pyrimidine, pyridine, pyrazole, quinoline, quinazoline) were applied. A distinct group of hybrids comprises the molecules built with natural compounds: Resveratrol, curcumin, coumarin, and oleanolic acid. In this review, we present the studies on bioactive hybrid molecules of a well-known tubulin polymerization inhibitor, combretastatin A-4 and its analogs with other pharmacologically active entities. The mechanism of anticancer activity of selected hybrids is discussed considering the structure-activity relationship.
Article
A series of novel 4,7-dihydroxycoumarin based acryloylcyanohydrazone derivatives were synthesized and evaluated for antiproliferative activity against four different cancer cell lines (A549, HeLa, SKNSH, and MCF7). Most of the compounds displayed potent cytotoxicity with IC50 values ranging from 3.42 to 31.28 µM against all the tested cancer cell lines. The most active compound, 8h was evaluated for pharmacological mechanistic studies on cell cycle progression and tubulin polymerization inhibition assay. The results revealed that the compound 8h induced the cell cycle arrest at G2/M phase and inhibited tubulin polymerization with IC50 = 6.19 µM. Experimental data of the tubulin polymerization inhibition assay was validated by molecular docking technique and the results exhibited strong hydrogen bonding interactions with amino acids (ASN-101, TYR-224, ASN-228, LYS-254) of tubulin.