Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

Received: 28 December 2019 Revised: 2 March 2020 Accepted: 2 April 2020

DOI: 10.1002/ptr.6703

REVIEW

Curcumin effect on cancer cells' multidrug resistance: An


update

Saeedeh Keyvani-Ghamsari1 | Khatereh Khorsandi2 | Asma Gul3

1
Department of Microbiology, Karaj Branch,
Islamic Azad University, Karaj, Iran Chemotherapy is one of the main methods for cancer treatment. However, despite
2
Department of Photodynamic, Medical Laser many advances in the design of anticancer drugs, their efficiency is limited due to
Research Center, Yara Institute, ACECR,
Tehran, Iran their high toxicity and resistance of cells to chemotherapeutic drugs. In order to
3
Department of Biological Sciences, improve the cancer therapy, it is essential to use the compounds that can overcome
International Islamic University, Islamabad,
drug resistance and increase treatment efficiency. Researchers have studied the
Pakistan
effects of natural compounds for the controlling various drug resistance mechanisms.
Correspondence
Curcumin is a natural phenolic compound which shows potent anticancer activities in
Khatereh Khorsandi, Department of
Photodynamic, Medical Laser Research Center, different tumors, alone or as an adjuvant with other antitumor drugs to prevent or
Yara Institute, ACECR, Tehran, Iran.
inhibit the survival and cancer progression by various mechanisms. The role of cur-
Email: khorsandi.kh@ut.ac.ir
cumin in overcoming drug resistance was followed by reviewing different applica-
tions of curcumin in cancer therapy. Afterward, the clinical impacts of curcumin, role
of curcumin in decreasing drug resistance in different cancer cells and its mechanisms
were discussed. It has been demonstrated that curcumin regulates signaling path-
ways in cancer cells, reduces the expression of proteins related to drug resistance,
and increases the performance of antitumor drugs at various levels. Curcumin
reverses multidrug resistance mechanisms and increases sensitivity of resistance cells
to chemotherapy. This review mainly focuses on different mechanisms of drug resis-
tance and curcumin as a nontoxic natural substance to eliminate the effects of drug
resistance through modulation and controlling cell resistance pathways and eventu-
ally suggests curcumin as a potent chemosensitizer in cancers.

KEYWORDS

anticancer drug, cancer, chemosensitizer, curcumin, multidrug resistance

1 | I N T RO DU CT I O N Reddy, & Velmurugan, 2017). At the beginning of treatment,


patients respond to chemical agent but unfortunately, cancer cells
There are different cancer treatment techniques such as surgery, che- become resistant to antitumor drugs over time leading to reduction
motherapy, radiation therapy, immunotherapy, hormone therapy, of life quality and longevity of the patient. In fact, drug resistance is
targeted therapy, stem cell transplant, and precision medicine, that the main reason for the failure of chemotherapy (Pan, Li, He, Qiu, &
alone or in combination are used for cancer patients (Palumbo Zhou, 2016). Involved factors in the drug resistance are divided into
et al., 2013). Treatment for each person depends on the type of can- two major categories: genetic and epigenetic factors in cancer cells
cer and stage of its development. One of the types of cancer treat- that reduce the sensitivity of cells to drugs and factors that block
ment is chemotherapy which is defined as application of various drug the access of cancer cells to the drugs (Nikolaou, Pavlopoulou, Geo-
classes that could-kill cancer cells through different mechanisms rgakilas, & Kyrodimos, 2018). These factors arise due to several
(Malhotra & Perry, 2003). It can also affect the normal cells (Aslam mechanisms such as changes in the drug intake, increases efflux
et al., 2014; Chan & Ismail, 2014; Huang, Ju, Chang, Muralidhar due to overexpression of P-glycoproteins (P-gps), DNA damage

Phytotherapy Research. 2020;1–23. wileyonlinelibrary.com/journal/ptr © 2020 John Wiley & Sons, Ltd. 1
2 KEYVANI-GHAMSARI ET AL.

repair, modified drug metabolism, and inhibition of cell death 2 | T H E M E C H A N I S M S O F DR U G


(Vosooghi et al., 2014). The genetic and epigenetic diversity ulti- RESISTANCE
mately favors the varieties of the resistance pathways available,
depending on the cancer itself; and even from an individual to indi- 2.1 | Increasing of drug efflux
vidual (Hervouet, Cartron, Jouvenot, & Delage-mourroux, 2013).
Therefore, the ability to predict and overcome drug resistance leads One of the mechanisms which cause drug resistance is the excessive
to development of chemotherapy and increases the survival rate of efflux of the drug, which is carried out by ATP-binding cassette trans-
patients. Since the different drug groups target various pathways, porters (ABC transporters; Housman et al., 2014). This superfamily
the chances of resistance to them decreases by using combination exists in all creatures from prokaryotes to humans. The structure of
therapy (Bozic et al., 2013). For this purpose, during the past these transporters has several subunits; two highly conserved cyto-
decades, many natural products which have chemosensitivity prop- plasmic domains that bind to ATP with energy utilizing properties,
erties and derived from plants, animals, fungi, and microorganisms, result in the release of various substrates across the cell membrane,
have been identified for combination therapy (Hamed, Abdel-Azim, and two less conserved transmembrane domains (Wilkens, 2015). In
Shams, & Hammouda, 2019; Khazaei Koohpar, Entezari, fact, by pumping toxins and drugs out of the cell, prevent the accumu-
Movafagh, & Hashemi, 2015). Among phytochemicals, curcumin— lation of these substances in the cell and in normal conditions main-
due to its low toxicity, side effects, and influence on various tain cells from xenobiotics (Chang, 2003; Sarkadi, Homolya,
cancers—has invited the attention of scientists. Curcumin is a poly- Szakács, & Váradi, 2006). In humans, 48 members of the ABC genes
phenol compound extracted from the Curcuma longa plant and is have been seen that three members of them play a main role in
commonly used as a spice (Goel & Aggarwal, 2010; Khazaei multidrug resistance (MDR), (a) P-gp, (b) MDR-associated protein
Koohpar et al., 2015; Liu, Chen, Cui, & Zhou, 2005). It acts as an 1 (MRP1), and (c) breast cancer resistance protein (BCRP/ABCG2;
antioxidant with anti-inflammatory, wound-healing, antiseptic, and Figure 1). P-gp which found in both normal and cancerous cells, in the
anticancer effects (Vallianou, Evangelopoulos, Schizas, & intestinal epithelium, liver cells, colon, proximal tubule of the kidney,
Kazazis, 2015; Wilken, Veena, Wang, & Srivatsan, 2011). Curcumin and blood–brain barrier could hydrolyzes two ATP to pump each drug
has shown increased sensitivity of cancer cells to treatment by molecule or toxin from inside to outside the cell (Sarkadi et al., 2006).
targeting various stages of cancer including cell cycle regulation, And since it is present on the cells before chemotherapy is considered
induction of apoptosis, reduction of metastasis, and reversal of as intrinsic resistance (Klopfleisch, Kohn, & Gruber, 2016). P-gp trans-
innate or acquired cellular resistance (Nabekura, 2010; Panda, fers broad substrate including topoisomerase inhibitors, microtubule-
Chakraborty, Sarkar, Khan, & Sa, 2017). In the present review we targeted drugs, tyrosine kinase inhibitors (TKIs), glucocorticoids, and
comprehensively describe different mechanisms of cellular resis- xenobiotics. Tissues such as lung, breast, and prostate that do not pro-
tance (Zhang & Wang, 2017), and activity of curcumin as a chemo- duce MDR, become resistant to drugs through the expression of
preventive agent that could overcome different drug resistance MDR1 and BCRP (Ambudkar, Kimchi-Sarfaty, Sauna, &
pathways in cancer cells. Gottesman, 2003; Housman et al., 2014). The structure and function

F I G U R E 1 Structure and function of


the multidrug ABC transporters. In
humans, the three major types of
multidrug resistance (MDR) proteins
include members of the ABCB (ABCB1/
MDR1/P-glycoprotein), the ABCC
(ABCC1/MRP1, ABCC2/MRP2, probably
also ABCC3–6, and ABCC10–11), and the
ABCG (ABCG2/MXR/BCRP) subfamily.
Multidrug/xenobiotic ABC transporters
are in the plasma membrane and extrude
various hydrophobic and/or amphipathic
xenobiotics and metabolic products.
MDR1/Pgp transports hydrophobic
compounds (X), while MRP1 and ABCG2
can extrude both hydrophobic drugs and
intracellular formed metabolites, for
example, glutathione or glucuronide
conjugates (C-X). Reprinted with
permission from Sarkadi et al. (2006).
ABC, ATP-binding cassette [Colour figure
can be viewed at wileyonlinelibrary.com]
KEYVANI-GHAMSARI ET AL. 3

of MDR1 is similar to P-gp, but it is GSH-dependent and only trans- systems depending on the type of lesion. DNA repair systems can be
fers drugs that are modified by glutathione (GSH). Therefore, it influ- categorized as base excision repair (BER), nucleotide excision repair
ences when the drugs are metabolically activated. MDR1 plays a key (NER), and mismatch repair (MMR) in single-strand damages and non-
role in the development of resistance to drugs such as etoposide, vin- homologous end joining (NHEJ), microhomology-mediated end joining
cristine, doxorubicin, epirubicin, vinblastine, and vincristine (MMEJ), and homologous recombination (HR) in double-strand breaks
(Liu, 2009). The BCRP structure is a homodimer composed of two half (Salehan & Morse, 2013). For example, cisplatin induces crosslinks in
transporter which are linked by disulfide bond. Mitoxantrone, doxoru- DNA structure that can cause cell death on the contrary, the resistant
bicin, topotecan, irinotecan, flavopiridol, and epirubicin can be men- cells escape from the damage with different repair mechanisms such
tion as BCRP specific substrates (Liu, 2009; Robey, Polgar, Deeken, as NER and HR (Rocha, Silva, Quinet, Cabral-Neto, & Menck, 2018;
To, & Bates, 2007; Sarkadi et al., 2006). Generally, overexpression of Figure 2). Studies on chronic myeloid leukemia cells have shown rate
these transporters in tumor cells has been observed to reduce the of O6-ethylguanine elimination and repair in patients that are resis-
accumulation of cytotoxic drugs in the cells which leads to cancer cell tant to alkylated drugs is higher than drug-sensitive patients (Müller,
survival and the development of MDR (Ambudkar et al., 2003). Thomale, Rajewsky, & Seeber, 1998). Moreover, in genotoxic stress, if
DNA cannot be repaired, DNA damage can be tolerated by specialized
DNA polymerases in the resistance cells. These DNA polymerases
2.2 | Increase in DNA damage repair have flexible and larger active site that cause the replication pass
through the lesions by the translesion synthesis (TLS) pathway and
Chemotherapeutic drugs, whether directly or indirectly, target the ultimately leads to mutation and drug resistance(Lehmann, 2005; Shi-
DNA. The change in DNA structure reduces replication and transcrip- mizu et al., 2003). Studies have suggested that several specific poly-
tion of the genes and eventually induces apoptosis in cells through merase enzymes are used in this system, one or two enzymes for
various pathways. One of the drug resistance mechanisms in cancer is nucleotides addition and the other enzymes for expansion (Salehan &
the removal of DNA damage that eliminates drug response and causes Morse, 2013). Overexpression of these polymers and DNA damage
tumor cells to survive (Stover, Konstantinopoulos, Matulonis, & recognition proteins has been observed in various cancer cells
Swisher, 2016). In these cells, the DNA damage is identified by DNA (Salehan & Morse, 2013; Srivastava et al., 2015) Studies reveal that
damage recognition proteins which activate a variety of DNA repair the overexpression of polymerase η reduces the susceptibility of

F I G U R E 2 Cisplatin-induced DNA
lesions and repair mechanisms that is
included: (a) The nucleotide excision
repair (NER) pathway for removing
cisplatin-induced DNA adducts (such as
1,2 and 1,3 intrastrand adducts), while the
mismatch repair (MMR) pathway can
recognize but not repair these adducts.
(b) Double-strand breaks are repaired by
homologous recombination (HR) and
nonhomologous end joining (NHEJ). The
NER, HR, translesion synthesis (TLS), and
Fanconi anemia (FA) pathways are
involved in ICL repair. (c) Schematic
representation of lesion bypass of
cisplatin-induced intrastrand crosslinks via
the TLS pathway. Reprinted with
permission from Rocha et al. (2018)
[Colour figure can be viewed at
wileyonlinelibrary.com]
4 KEYVANI-GHAMSARI ET AL.

ovarian cancer stem cells (CSCs) to cisplatin so that downregulation of modifications of ER in breast cancer cells which prevent the binding
Pol η increased apoptosis in cells treated with cisplatin (Srivastava of tamoxifen to ER could be consider as one of resistance mechanisms
et al., 2015). Another study has shown that expression of polymerase to tamoxifen (Criscitiello, Fumagalli, Saini, & Loi, 2011; Likhite, Stossi,
η increases in gemcitabine and cisplatin-resistant head and neck squa- Kim, Katzenellenbogen, & Katzenellenbogen, 2006).
mous cancer cell (Zhou et al., 2013). Tyrosine kinase inhibitors target the epidermal growth factor
receptor (EGFR) in the cells which generally their use fails after about
1 year. One of the main reasons for their resistance is mutations in
2.3 | Altering the drug targets the kinase domain of EGFR genes. So, by mutations in the gatekeeper
residues the binding of drug to catalytic ATP-binding site in the kinase
During drug targeting, a drug often encounters resistance mecha- domain of EGFR is prevented (Bell et al., 2005).
nisms, such as modification of the special target of the drug through
mutation or alteration in the level of expression of its target which
results in a change in the quantity or quality of the drug target 2.4 | Resistance to apoptosis
(Alfarouk et al., 2015). For example, taxanes cause cancer cell death
through binding to tubulins. Reviews have shown the change in the Apoptosis is programmed cell death through intrinsic and extrinsic
structure of tubulins due to gene mutation or inactivation of tubulin- pathways under cellular stress or receiving death signals from other
specific chaperons is a major cause of tubulin resistance in breast can- cells. In the external pathway, binding of ligands to death receptors at
cer. In addition, downregulation of tumor β III- and β V-tubulins has the cell surface activates caspases 8 and 10 that cleave and activate
been observed in the resistant cells (Nami & Wang, 2018). Irinotecan effector caspases 7, 6, and 3 (Fulda & Debatin, 2006). Internal path-
binds to DNA topoisomerase I complex and through creating perma- way performed in the mitochondria, with creating apoptosome by
nent cleavages in DNA structure, inhibit reproduction and transcrip- cytochrome c, Apaf-1, and procaspase 9 that activates caspase 9, 7,
tion. The topoisomerase I expression reduced in colon cancer 6, and 3 (Pfeffer & Singh, 2018). In this process, the balance between
resistance cells which reduced the sensitivity of the cells to the drug antiapoptotic proteins, such as Bcl2, Bcl-Xl, and AKT and the
(Boyer et al., 2004; Pommier, 2013). Tamoxifen is a selective estrogen preapoptotic proteins such as Bax, Bak, and Bid leads to apoptosis
receptor (ER) modulator which is used for the treatment of breast can- (Baig et al., 2016; Figure 3). The raising of antiapoptotic factor expres-
cer. Mutation in ER gene, its downregulate, and posttranslational sion and reduction the proapoptotic factor expression play a key role

F I G U R E 3 Schematic representation
of the extrinsic and intrinsic apoptotic
pathway which could be induced by
chemotherapy and radiotherapy drugs.
The increasing of antiapoptotic factor
expression (IAPs) and reduction the
proapoptotic factor expression (Bax, Bak,
and Bid) play a key role in progress of the
drug resistance. Reproduced with
permission from de Vries (2006). IAP,
Inhibitors of apoptosis [Colour figure can
be viewed at wileyonlinelibrary.com]
KEYVANI-GHAMSARI ET AL. 5

in progress of the drug resistance (Razaghi, Heimann, Schaeffer, & system promote inactivation reactions of drugs and protects cells
Gibson, 2018; Siddik, 2003). Therefore, targeting these proteins is an against the chemotherapy. Additionally, the drug binding to GSH
important factor in cancer treatment. The use of inhibitors of anti- increases the solubility of the drug and enhances the possibility of
apoptotic proteins such as antisense oligonucleotides, antibodies, or drug exit from the cell by MDR proteins (Wang, Yang, et al., 2015).
small molecule inhibitors induce apoptosis in the tumor cells and The UGT (also called glucuronosyltransferase) family is a part of Phase
enhance the effect of chemotherapy drugs, but studies have shown II metabolism that is responsible for glucuronidation to various sub-
their long-term use also causes resistance (Garc, Elisabet, & strates such as bilirubin, bile acids, steroids, and drugs. UGT1 and
Redondo, 2018; Soria et al., 2010). Inhibitors of apoptosis (IAPs) as UGT2 are found in human that have important role in the body's met-
caspase inhibitors, also have remarkable role in the drug resistance abolic defense system. It is reported epigenetic changes reduce the
(Razaghi et al., 2018). Overexpression of X-linked IAP (XIAP) has been expression of these genes in some cancers. In fact, DNA promoter
seen in cancer cells whose its inhibition by short-interfering RNA methylation of these genes leads to gene silencing that drugs such as
(siRNA) enhanced apoptosis in the cells and increased the effect of irinotecan are active in these conditions. However, studies have
chemotherapy on the cells (Wong, 2011). In addition, P53 is a tumor shown overexpression of these genes causes resistance to irinotecan
suppressor protein cods by TP53 gene which effects the transcription and other drugs (Holohan, Van Schaeybroeck, Longley, &
of the responsible genes in apoptosis and leads to apoptosis at the Johnston, 2013; Pathania, Bhatia, Baldi, Singh, & Rawal, 2018).
time of DNA damage. Mutation and losing in Tp53 gene results in cell
proliferation despite gene damage in cancer cells. The mutation of this
gene occurs in various cancers in the range of 10–100%, this action 2.6 | Role of epigenetic changes in MDR
leads cancer cells ultimately resistance to the chemotherapy (Geisler
et al., 2003; Rivlin, Brosh, Oren, & Rotter, 2011; Talib, Al-hadid, Wild Epigenetic modifications are another mechanism of drug resistance in
Ali, Al-Yasari, & Abd Ali, 2018). cancer treatment (Fardi, Solali, & Farshdousti Hagh, 2018). DNA
methylation and histone modification (acetylation or methylation) are
two main types of the epigenetic alterations directly affecting the pro-
2.5 | Decreasing the activity of the drugs moter of genes or indirectly alter chromatin conformation (Kanwal &
Gupta, 2012; Figure 4). These modifications alter the cell phenotype
Antitumor drugs could react with the proteins and enzymes in the by affecting gene expression without altering the genotype
cells and be activate after structural changes. Changes to drug activa- (Miremadi, Oestergaard, Pharoah, & Caldas, 2007). More recently,
tion include modification, degradation, or the binding of drugs to research has shown that these changes can lead to acquired drug
other molecules. But generally after beginning treatment, the drug resistance. For example, hypomethylation in promoter regions of
activation process in cancer cells is impaired and the toxic effect of oncogenes, upregulates the gene expression, and hypermethylation
the drug is reduced (Zahreddine & Borden, 2013). Cytochrome P-450 reduces expression of tumor suppressor genes in cancer cells
(CYPs) and glutathione S-transferase (GST)/glutathione S-hydrolases (Baker & El-Osta, 2003). Modification of histones also results in relax-
(GSH) and the uridine diphospho-glucuronosyltransferase (UGT) ation or condensation of chromatin, thereby affecting gene expres-
superfamily could be mention as important enzymes for activating sion. For example, H3K9 methylation at the promoter of many tumor
and inactivation of drugs (Michael & Doherty, 2005). The cytochrome suppressors causes gene silencing. Whereas, histone acetylation gen-
P-450 superfamily enzymes are present in the inner membrane of erally cause chromatin to open and transcription factors can access
mitochondria or in the endoplasmic reticulum in the most tissues of DNA-encoded regions (Hu & Baeg, 2017). Studies have shown inap-
the body and have a wide range of substrates such as xenobiotic, ste- propriate methylation of the ER promoter, histone deacetylation, and
roids, cholesterol, and hormones (P. B. Danielson, 2002). They are hypoxia decreases receptor expression and leading to cell resistance
Phase 1 enzymes in the metabolism of the most drugs such as cyclo- to tamoxifen (García-Becerra, Santos, Díaz, & Camacho, 2013). Other
phosphamide, epipodophyllotoxins, ifosfamide, teniposide, tamoxifen, studies have reported methylation at the MDR1 gene promoter regu-
taxol, and vinca alkaloids. The mutation in cytochrome p450 genes, its lates the expression of this protein. Demethylation of MDR1 pro-
deficiency, and inhibition is responsible for the inactivation of these motor upregulates expression of the protein in the resistance cells
drugs (Bibi, 2008; Talalay, 2000). The GST–GSH system is known as which reduces the accumulation of drugs inside the cell, whereas the
Phase II metabolic isozymes that play an important role in the mecha- promoter of this gene is hypermethylated in drug-sensitive cells
nism of detoxification of drugs by catalyzing the conjugation of GSH. (Baker & El-Osta, 2003; Klopfleisch et al., 2016). The Epigenetic
They are potent antioxidant that protects cells against toxic com- changes also affect the expression of DNA repair enzymes.
pounds, free radicals, and peroxidases. Although they have a positive Hypermethylation in the promoter of MMR enzymes including the
effect on the protection of the normal cells against reactive oxygen human MutL homolog 1(hMLH1) and human MutS protein homolog
species (ROS) which cause resistance in chemotherapy (Allocati, Mas- 1 (hMSH1) result in gene silencing which can cause the development
ulli, Di Ilio, & Federici, 2018). The chemotherapeutic agents induce of cancers (Klopfleisch et al., 2016). Epigenetic changes are reversible
and increase the expression of these proteins (Talalay, 2000; Wang, by DNA methyl transferase (DNMT) inhibitors and histone
Yang, Yang, Wang, & Lee, 2015). Increasing the activity of GST–GSH deacetylase (HDAC) inhibitors. For example, the epigenetic
6 KEYVANI-GHAMSARI ET AL.

F I G U R E 4 Three different epigenetic levels: DNA methylation, histone methylation or acetylation, and chromatin reading. The use of
demethylating agents and HDAC inhibitors offers the potential to change the gene expression profile of tumors lead to tumor cell death and
increased apoptotic response to chemotherapy drugs. Reproduced with permission from Jubierre et al. (2018). HDAC, histone deacetylase
[Colour figure can be viewed at wileyonlinelibrary.com]

modifications in hMLH1 and inactivation of this gene can lead to can- upregulation of the miR- 206, miR-106a, miR-21, and miR-214 in
cer progression and resistance to drugs such as cisplatin, carboplatin, these cells reduced the expression of ERa, RB1, and PTEN proteins
temozolomide, epirubicin, and 5-fluorouracil. However, the use of and resulting in resistance to doxorubicin. Study on the expression of
DNMT inhibitors such as decitabine and azacytidine return the sensi- MDR1 protein in MCF-7 cells has shown that the expression of this
tivity of cancer cells to drugs by reducing methylation of the hMLH1 protein is inversely related to expression of miR-451. Transfection of
promoter (Klopfleisch et al., 2016; Mansoori, Mohammadi, Davudian, these cells with miR-451 decreased MDR1 expression and therefore
Shirjang, & Baradaran, 2017). increased cell susceptibility to doxorubicin (Kovalchuk et al., 2008).
Another study has shown miR-7 (as a tumor suppressor agent in vari-
ous cancers) downregulates in cetuximab-resistant cells. On the other
2.7 | MicroRNA and drug resistance hand, increasing miR-133b in the tumor cells with the effect on EGFR
suppressed the growth of cells and increased the sensitivity of malig-
MicroRNAs (miRNAs) are small molecules of about 21–25 nucleotides nant cells to cetuximab (Ghasabi et al., 2019). Upregulation of miR-
that are unable to encode a protein instead play a key role in posttran- 27a, miR-451, miR-99a, miR-100, and miR-125b1 was seen in the
scriptional regulating of gene expression (Ghasabi et al., 2019). This drug-resistance ovarian and cervical cancer cells. The use of miR-27a
large family affects the expression of about 60% of genes encoding or miR-451 antagonists reduced P-gp and MDR1 at the cell surface
proteins. MiRNAs are coupled to their complementary base and consequently increased the concentration of the drugs inside the
sequences in the 30 region of mRNAs, and through several mecha- cells (Ma, Dong, & Ji, 2010). Studies on tamoxifen-resistant breast
nisms silence mRNAs (Fabian, Sonenberg, & Filipowicz, 2010; Ghasabi cancer cells have shown that about eight miRNAs upregulate and
et al., 2019). These mechanisms include fragmentation of the mRNA seven miRNAs downregulate in the cells compared with tamoxifen-
into two separates pieces, shorten the poly (A) tail in the mRNA struc- sensitive cells (Miller et al., 2008). Since microRNAs can affect the
ture that making it unstable, and block protein translation through the advancement of various stages of resistance, pay attention to them
ribosome. Therefore, they epigenetically control the expression of can help to overcome the drug resistance (Figure 5).
genes such as tumor suppressors genes or oncogenes (Jalkanen, Col-
eman, & Wilusz, 2014). Recent studies have proven that miRNA dys-
regulation promotes the drug resistance in various cancers (Kumar, 2.8 | Role of CSCs in drug resistance
Kushwaha, & Gupta, 2019). For example, downregulation of miR-127
and miR-34a reduced apoptosis in the MFC-7 cells treated with doxo- Cancer stem cells have been seen in hematopoietic and solid tumors.
rubicin by decreasing p53 signaling pathway. On the other hand, They are capable of self-renewal and differentiation and play an
KEYVANI-GHAMSARI ET AL. 7

F I G U R E 5 Different Mechanisms of
multidrug resistance in cancer cell could
be classified into many different
mechanisms such as enhanced drug
efflux, enhanced DNA damage repair,
altering drug targets, inhibiting apoptosis,
decreasing the drug activity, cell cycle
checkpoints alternation, and epigenetics
alternation. MiRNAs could mediate drug
responses of cancer cells by targeting the
pathways related to resistance. Reprinted
with permission from Zhang and
Wang (2017) [Colour figure can be
viewed at wileyonlinelibrary.com]

important role in tumor initiation, invasion, metastasis, and drug resis- cell's self-renewal. Signaling pathways activated by hypoxia also
tance (Najafi, Mortezaee, & Ahadi, 2019; Saha, Adhikary, decrease ROS production in CSCs, and by creating a state of silence in
Bhattacharyya, Das, & Sa, 2012). CSCs are a small population of cells the cells develop resistance to treatment (Prieto-Vila et al., 2017;
within a tumor that mainly located in a special environment called the Zhang et al., 2018). CSCs also have protective autophagic mechanisms
niche with hypoxia, low pH, and low nutrition (Mondal, that maintain their stability (You et al., 2019). MiRNAs are another
Bhattacharya, & Mandal, 2018). They are surrounded by a variety of factor that through epigenetic changes leads to the survival of CSCs,
cells such as fibroblasts, endothelial, mesenchymal, and immune cells their stemness and consequently drug resistance (Najafi, Mortezaee, &
in the niche that promote self-renewal of CSCs (Prieto-Vila, Taka- Majidpoor, 2019). Therefore, given the importance of CSCs in tumor
hashi, Usuba, Kohama, & Ochiya, 2017). According to one theory, maintaining and MDR in the treatment, targeting, and eliminating
CSCs may be caused by genetic or epigenetic alterations of normal them helps to overcome drug resistance.
stem cells. Another theory suggested that the origin of CSCs is muta-
tion in differentiated cancer cells (Mondal et al., 2018). Many studies
have shown that CSCs use both intrinsic and extrinsic resistance 3 | NA T U R A L P R O D U C T S OV E R C O M E
mechanisms to escape death against antitumor drugs and exhibit M D R I N C A N CE R C E L LS
more resistance to chemotherapy than non-CSCs. Due to their unlim-
ited self-renewal ability, they can regenerate tumor cells after chemo- Since different parts of the cell are targeted in cancer, treating the dis-
therapy (Najafi, Mortezaee, & Majidpoor, 2019). Among the ease with one drug is almost impossible. In addition, the cells quickly
mechanisms of resistance in CSCs, overexpression of ABC trans- find ways to escape the mechanisms of drug functional and adapt to
porters such as ABCB1, ABCG2, ABCB5, and ABCC1 on the surface therapy. The discovery of new drugs with high efficacy and low toxic-
of these cells could be mentioned (Prieto-Vila et al., 2017). They also ity that have selectively is one of the goals of cancer treatment. In
avoid apoptosis by overexpression of antiapoptotic proteins such as recent decades, the use of natural compounds has attracted the atten-
Bcl-2, surviving, and Mcl-1 or through disable cell cycle regulators. tion of scientists in cancer therapy (Nobili et al., 2009). These com-
The DNA damage–repair system is also highly active in these cells pounds have been used for centuries to treat a variety of diseases and
(Najafi, Mortezaee, & Majidpoor, 2019; Tsai et al., 2012). Studies have are taken from various parts of the plant tissues. Currently, many
shown that epithelial–mesenchymal transition (EMT) regulators are patients from Australia, Europe, and the United States are using herbal
very effective in these cells. Due to the close association of EMT to compounds in their combination therapies (Ekor, 2014). Natural com-
the biology of CSCs and the role of EMT on non-CSCs to transform pounds due to their high efficacy and low toxicity and chemo-
into CSCs, it is referred to another resistance factor in these cells sensitizing activities are quite convenient in combination therapy
(Najafi, Mortezaee, & Ahadi, 2019; Wang, Sun, Provaznik, Hackert, & (Aung, Qu, Kortschak, & Adelson, 2017). So far only about 10% plant
Zöller, 2019). The next mechanism that causes resistance in CSCs is species have been identified for the treatment of various diseases.
the hypoxic position of the malignant stem cells which is the provider Among natural compounds flavonoids, alkaloids, steroids, monoter-
of growth signals, transformation, and maintaining the stability of the penes, limonoids, saponins, polyphenols, and coumarins as anticancer
8 KEYVANI-GHAMSARI ET AL.

compounds could be mentioned (Eid, El-Readi, Fatani, Mohamed Nour suppressing tumor progression (Figure 6). This findings show curcumin
Eldin, & Wink, 2015; Iqbal et al., 2017; Millimouno, Dong, Yang, Li, & can increase susceptibility of cancer cells to chemotherapy. Also, cur-
Li, 2014). Phytochemicals could active or inhibit different enzymes in cumin is used alone or in combination with other drugs to treat vari-
cell proliferation, apoptosis, oncogenes, DNA repair signaling path- ous types of cancers such as breast, colon, head and neck cancer,
ways and many of them act as chemopreventive agents (Nabekura, melanoma, pancreas, and lung (Panda et al., 2017).
Yamaki, Ueno, & Kitagawa, 2008). Flavonoids such as quercetin,
morin, citrus, curcumin, and capsaicin inhibit the drug efflux by the
inhibition of ABC transporters, MDR1, MRP, and BCRP with directly 4 | C U R C U M I N O V E R C O M ES D R U G
connecting to the ATP-binding sites and in this way overcome MDR RESISTANCE
(Conseil et al., 2002). Citrus flavonoids not only affect ABC trans-
porters but also regulate the function of metabolic enzymes (GST and As previously described, the main problem in cancer treatment and
P450) in activation of chemotherapy agents (Eid et al., 2015). chemotherapy is the toxicity of drugs in the normal cells and the resis-
Epigallocatechin gallate in tea, curcumin in turmeric, and capsaicin in tance of cancer cells to the mechanism of action of drugs that thereby
chili pepper reduce P-gp at the surface of human carcinoma KB-C2 evade apoptosis. Numerous studies have demonstrated the ability of
cells in a concentration-dependent behavior, cause drug accumulation curcumin to resolve drug resistance. There are some purposed mecha-
within the cells and increase the sensitivity of these cells to daunoru- nisms for curcumin ability to overcome drug resistant that could be
bicin and vinblastine (Kitagawa, Nabekura, & Kamiyama, 2004; described as below.
Nabekura, 2010). Berberine through removing the MDR in K562/A02
cells increases cell sensitivity to adriamycin and doxorubicin (Meng,
Ze-Fa, Hua, & Bao-An, 2018). 4.1 | Inhibition of drug efflux

Lack of accumulation of drugs within the cancer cells due to over-


3.1 | Curcumin expression of ABC transporters plays an important role in MDR. Cur-
cumin inhibits ABC family transporters such as ABCA1, ABCB1,
Among phytochemicals, curcumin has been considered because of its ABCC1, and ABCG2 in various cancer cells. For example, in cervical
potent anticancer properties and minimal side effects. Curcumin is an cancer cells curcumin reduces the expression of ABCB1 (P-gp) protein
active polyphenolic pigment of turmeric derived from Curcuma longa at the surface of cells as well as by interacting with the P-gp binding
rhizomes, generally grows in tropical logic, especially in Southeast Asia sites, significantly decreasing the activity of this transporter (Table 1).
that its use dates back to about 5,000 years ago that was generally Treatment of these cells with vinblastine plus curcumin leads to accu-
used as an inflammation reducing agent in infection and autoimmune mulation of vinblastine within the cells and increases the sensitivity of
diseases (Panda et al., 2017; Pongrakhananon & Rojanasakul, 2012). It the cells to the drug (Limtrakul, Anuchapreeda, & Buddhasukh, 2004;
is known for its antioxidant, anti-inflammatory, wound-healing, and Zhang, Chen, Wang, Peng, & Cai, 2014). Research has shown cur-
anti-carcinogenesis properties that prevent the beginning or develop- cumin to reduce ABCG2 function, increases the sensitivity of tumor
ment of the cancer (Khorsandi et al., 2015; Krausz et al., 2015). One cells expressing this transporter to drugs such as, SN-38, doxorubicin,
of the major problems of using curcumin is its poor solubility, low topotecan, and mitoxantrone (Chearwae, Shukla, Limtrakul, &
absorption, and limited tissue distribution. Encapsulation of curcumin Ambudkar, 2006). Others have also shown combination of 5 mg doxo-
in liposome, micelles, silica, intercalated into cyclodextrine rubicin and 5 mg curcumin in nanoparticle formulation increases
nanoparticles or into layered double hydroxide (LDH) nanohybrids are doxorubicin retention in the nucleus and enhances doxorubicin toxic-
one of the few solutions (Khorsandi et al., 2015; Khorsandi, ity in K562 cells. Curcumin inhibits MDR1, Bcl-2, and BCR/ABL gene
Hosseinzadeh, & Shahidi, 2019) that remove the drug restrictions and expression in these cells and by eliminating cellular resistance
enhance the effectiveness of the curcumin (Khorsandi et al., 2019; enhances doxorubicin efficacy in the cells (Misra & Sahoo, 2011).
Krausz et al., 2015; Rai, Pandit, Gaikwad, Yadav, & Gade, 2015). Cur- Therefore, curcumin is a useful agent in combination with important
cumin inhibits ABC transporters, regulates production of metabolic chemotherapy drugs to overcome MDR.
enzymes Phase I and II, downregulates genes that respond to DNA
damage such as ATM, ATR, BRCA1, and O6-methyl guanine-DNA
methyl transferase (MGMT) in cancer cells (Mori et al., 2006; Saha 4.2 | Induction of DNA damage
et al., 2012). Microarray studies have shown that, the expression of
104 genes changes in the apoptotic signaling pathways with the pres- Another factor in drug resistance was the inhibition of DNA damage
ence of curcumin. In addition to apoptosis, curcumin affects the regu- caused by antitumor drugs. There are many evidences in relation to
lation of metastatic, angiogenesis, cell cycle checkpoints, and immune the inhibition of DNA repair special enzymes by curcumin and also
system signaling pathways (Ramachandran et al., 2005; Saha curcumin induces DNA damage in different cell lines (Lu et al., 2009;
et al., 2012). In general, main anticancer activities of curcumin include Rowe, Ozbay, O'Regan, & Nahta, 2009; Figure 7). Real-time results in
inhibition of tumor formation, induction of apoptosis death, and mouse–rat hybrid retina ganglion cell line N18 showed that 20 μM of
KEYVANI-GHAMSARI ET AL. 9

F I G U R E 6 Main anticancer
activity of curcumin such as
inhibition of tumor formation,
induction of apoptosis death, and
suppressing tumor progression
[Colour figure can be viewed at
wileyonlinelibrary.com]

TABLE 1 Curcumin and its derivatives with P-gp modulatory effect

Inhibitors of Inhibitors of
Compounds MDR cancer model P-gp function P-gp expression References
Unsymmetrical curcumin mimics MDR cervical adenocarcinoma X (Um et al., 2008)
with various amide moieties
Heterocyclic cyclohexanone Human embryonic kidney cells and X (Revalde, Li, Hawkins,
monocarbonyl analogs of canine kidney cells transfected Rosengren, &
curcumin with wild-type P-gp Paxton, 2015)
Chloro and asymmetrical series of MDR acute lymphoblastic leukemia X (Ooko et al., 2016)
synthetic curcumin derivatives
Diketone and cyclohexanone MDR chronic myeloid leukemia X X (Mapoung et al., 2016)
curcumin analogs

Abbreviation: MDR, multidrug resistance.

curcumin for 48 hr decreases the expression of DNA damage–response HR pathway. Curcumin downregulated the expression of Rad51 and
genes such as; ATM, ATR, BRCA1, DNA-PK, and MGMT (Lu et al., 2009). removed the resistance of the cells to the drugs which ultimately lead to
Other studies have shown that curcumin induces DNA damage, modu- the induction of caspase3-dependent apoptosis (Zhao et al., 2018).
lates BRCA1, and maintained this protein within the cytoplasm in triple-
negative breast cancer cells, loss of BRCA1 in nucleus prevents the repair
of the DNA and induces apoptosis in these cells (Rowe et al., 2009). In 4.3 | Disable epigenetic changes
one study, it was shown that curcumin inhibits the FA/BRCA pathway in
cyclophosphamide resistant human lymphoma cell line HT/CTX. As As mentioned earlier, epigenetic changes play an important role in the
FA/BRCA pathway regulates the cellular response to DNA damage, cur- development of drug resistance in cells. There are many reports in
cumin reverses the cyclophosphamide resistance in HT/CTX cells and relation to suppress DNMT by curcumin.
increases the drug toxicity in these cells (Ji, 2010). In CH12F3 lymphoma Studies on binding of curcumin to DNMT1 via molecular docking
cells, 5 μM of curcumin sensitized cells to various DNA damage drugs have shown that curcumin binds covalently to the catalytic active site
including cisplatin, methyl methanesulfonate, hydroxyurea, and camp- of DNMT1 and blocks it (Liu et al., 2009). Another study has shown
tothecin. In these cells curcumin-induced DNA break through regulating 10 μM of curcumin inhibits DNMT1 in acute myeloid leukemia (AML),
Rad51-dependant HR. HR is a main pathway in repairing double-strand both in vitro and in vivo. In addition, it downregulates the positive
breaks induced by antitumor drugs and Rad51 is an essential factor in regulators of DNMT1, including p65 and Sp1 which reduces the
10 KEYVANI-GHAMSARI ET AL.

methylation through inhibition of DNMT3b by curcumin increased


expression of phosphatase and tensin homolog (PTEN), which plays
an effective role in inhibiting cell proliferation and inducing apoptosis
in hepatic stellate cells (Zheng et al., 2014). In addition to DNA meth-
ylation, acetylation of histones plays key role in gene expression
changes. Therefore, HDAC inhibitors are considered in the treatment
and enhancement of cell response to the chemotherapy (Reuter,
Gupta, Park, Goel, & Aggarwal, 2011). Bora-Tatar et al. have shown
that μM doses of curcumin has a potent inhibitory effect on HDAC, as
its effect is stronger than the known HDAC such as valproic acid and
sodium butyrate (Bora-Tatar et al., 2009). In B-NHL cell line Raji cells,
curcumin has reduced the expression of HDAC1, HDAC3, and
HDAC8 proteins thereby increasing the expression of acetylated his-
tone H4 and induces apoptosis (Liu et al., 2005). Another study has
F I G U R E 7 Curcumin-induced DNA damage and inhibition of shown that curcumin reduces expression of HADC4 in medulloblas-
DNA repair associated protein expression in human cervical cancer toma cells which leads to increased tubulin acetylation. Ultimately,
HeLa cells. Reproduced with permission from Shang et al. (2016)
inhibiting cell growth at the G2/M phase and inducing apoptosis in
the cells (Lee et al., 2011; Figure 8).
binding of these factors to the DNMT1 promoter. DNA hyp-
omethylation plays an important role in activating tumor suppressor
genes in AML, therefore proving curcumin a potent agent in the treat- 4.4 | Inhibition of metabolic enzymes
ment of AML (Boyanapalli & Kong, 2015). In hepatocellular carcinoma
that curcumin inhibits cell proliferation and induce apoptosis with As explained above, overexpression of Phase II metabolic isozymes
reduction in expression level of DNMT1 (Sanaei & Kavoosi, 2018). plays an important role in the inactivation of drugs within the cell, so
Another study in vivo and in vitro showed, reduction of DNA the use of their inhibitors can be effective in preventing the

F I G U R E 8 Curcumin disable epigenetic changes. Curcumin could alter DNA methylation, histone modifications, and miRNA activity.
Reproduced with permission from Hassan et al. (2019) [Colour figure can be viewed at wileyonlinelibrary.com]
KEYVANI-GHAMSARI ET AL. 11

development of drug resistance. Studies have shown that curcumin oral, MCF-7, and non-small cell lung carcinoma that limits the prolifer-
reduces glutathione transferases (GSTs) activity at both gene and pro- ation of cancer cells, causes cellular accumulation in the G1 phase and
tein levels. For example, in human IGR-39 melanoma cells, curcumin also increases the sensitivity of the cells to the drugs (Meiyanto
inhibited GST activity through covalent modification of the enzyme et al., 2014; Shishodia et al., 2005). Overexpression and continuous
and in K562 cells strongly reduced the expression of GST P1-1 mRNA NF-κB activity has been observed in human head and neck squamous
(Mathew, Kalyanasundaram, & Balaraman, 2006). Downregulation of cell carcinoma cell lines. It has been proven that curcumin suppresses
GSTP1-1 mRNA by curcumin in K562 cells is associated with inhibi- the activity of NF-κB in these cells through inhibition IκBα kinase
tion of TNF-α, AP-1, and NF-κB binding to the GST P1-1 gene pro- which is essential for the activation of NF-κB (Aggarwal, Takada,
moter (Duvoix et al., 2003). The effect of curcumin on Phase II Singh, Myers, & Aggarwal, 2004).
enzyme in rat liver is dose-dependent. Low concentrations of Curcumin also downregulates NF-κB expression by blocking the
curcumin-induced GST expression while high concentrations inhibited PI3K/AKT pathway. NF-κB inhibition directly affects P53 activation,
GST expression (Mathew et al., 2006). On the other hand, a number on the other hand, inhibits the expression of COX-2 and VEGF genes
of studies reported curcumin induces GST expression in the cells, thereby reducing angiogenesis in cancer cells. Studies have shown
which is effective in removing toxic and oxidizing substances and thus that the combination of 5-fluorouracil (5-FU) with curcumin signifi-
playing an important role in the prevention of cancer in early stages cantly reduces tumor growth in mice-bearing human gastric cancer
(Nishinaka et al., 2007; Syng-Ai, Kumari, & Khar, 2004). The effects of xenografts compared with the use of 5-FU alone. The results showed
curcumin on cytochrome P450 2C9 (CYP2C9) in human and cyto- this combination reduces the level of NF-κB and COX-2 protein by
chrome P450 2C11 (CYP2C11) in rat liver microsomes revealed that 37 and 45%, respectively. In fact, curcumin suggested the well-known
curcumin inhibits the activity of these cytochromes in a noncom- natural compound that inhibited NF-κB activity and increased the
petitively manner for the enzyme substrate and uncompetitive for the sensitivity of cancer cells to treatment (Mortezaee et al., 2019). Cur-
cofactor NADPH. Given the role of CYP2C9 in activating a number of cumin also inhibits cyclin D1 activity through NF-κB inhibition as seen
the drugs, more attention should be paid when curcumin combined in head and neck squamous carcinoma (Aggarwal et al., 2004). Arrest-
with drugs that are metabolized by CYP2C9 (Wang et al., 2015). The ing the growth of cancer cells at various stages by curcumin can be
use of curcumin in rats has shown that curcumin decreases P-gp and one option to overcome MDR.
activates CYP 3A4 in vivo and ex-vivo, since CYP 3A4 is required to
metabolize more than 50% of drugs and also has a positive role in
activating multiple drug–drug interactions, curcumin can be a useful 4.6 | Regulation of miRNAs
agent for the activation of certain medicines (Hsieh et al., 2014).
MiRNAs dysregulates in many tumors and through the influence on
the different molecular and cellular pathways such as NF-κB, protein
4.5 | Curcumin effect on NF-κB expression kinase B, VEGF, MAPK, and cell cycle play an important role in the
proliferation, apoptosis, migration, and the resistance of cells to che-
There are many reports about the inhibitory effect of curcumin on motherapy (Boyanapalli & Kong, 2015; Mirzaei et al., 2018). Several
inflammatory activities in various tumors through NF-κB inhibition studies have shown that curcumin regulates miRNAs expression such
(Escárcega, Fuentes-Alexandro, García-Carrasco, Gatica, & as miR-1, miR-7, miR-9, miR34a, miR-21, miR-27, miR-181, and miR-
Zamora, 2007). NF-κB is a nuclear transcription factor that has a key 186 thereby inhibiting proliferation of cancer cells and also increases
role in the cell proliferation, cell survival, and inflammatory responses. the sensitivity of cells to antitumor drugs (Mirzaei et al., 2017).
It is required for the expression of cytokines, growth factors, onco- Research on cloned cancer cells has shown that expression of miR-21
genes, and cell cycle regulators (Spiros, 2017; Xia et al., 2018). NF-κB is increased in these cells which induces cell proliferation and metas-
regulates the expression of responsible genes for the immune system tasis via reducing the expression of several proteins. One of the tar-
and inflammation. The potential connection between chronic inflam- gets of miR-21 is the tumor suppressor PDCD4 that downregulated in
mation and the initiation of cancer has been proven (Escárcega the presence of miR-21. The effect of 5–20 μM of curcumin for 24 hr
et al., 2007; Jacob, Wu, Zhou, & Wang, 2007). on clonal cancer cells has shown that curcumin significantly down-
Improper regulation of NF-κB has been seen in many types of regulates miR-21 in primary tumors and also inhibits cell growth in
cancer, moreover it increases in the presence of most anticancer invasive and metastatic cells (Mudduluru et al., 2011). Another miR-
drugs in the cancer cells. NF-κB induces antiapoptotic proteins and 21 target is tumor suppressor protein PTEN, which inhibits cell prolif-
rises MDR protein expression that leads to cell viability and cellular eration by affecting the cell cycle. One study has shown that
resistance in cancer cells (Xia et al., 2018). Curcumin inhibits constitu- difluorinated-curcumin (CDF) one of the curcumin derivatives
tive NF-κB activity and blocks NF-κB binding to the DNA whereby decreases the expression of miR-21 in chemoresistant cells of colon
reduces the expression of genes regulated by NF-κB such as TNF, cancer thereby increased the PTEN level and suppressed the drug-
IkBa, Bcl-2, Bcl-xl, COX-2, and IL-6 genes in various cancer cells resistant cells proliferation (Roy, Yu, Padhye, Sarkar, &
(Shishodia, Amin, Lai, & Aggarwal, 2005). Downregulation of NF-κB in Majumdar, 2013). Studies on breast cancer cells have shown that cur-
the presence of curcumin has been observed in mantle cell lymphoma, cumin upregulates miR-15a and miR-16 in these cells, which both of
12 KEYVANI-GHAMSARI ET AL.

them inhibits the expression of Bcl-2. As a result, depletion of this anti- BCSC population also reached about 25% of untreated cells. In addi-
apoptotic protein induced apoptosis in the cells (Yang, Cao, Sun, & tion, curcumin increased the sensitivity of cancer cells to mitomycin
Zhang, 2010). Microarray analysis has shown that curcumin upregulates C. This combination deregulated Bcl/Bax complex, increased
expression of miR-22, miR-200c, let-7 g*, and miR-923 which are caspase-3 and caspase-9 activity thus increased apoptosis in BCSCs
tumor suppressor and downregulates expression of 16 miRNAs in reti- (Liu & Ho, 2018).
noblastoma cells, ultimately reducing cell proliferation and migration
(Sreenivasan, Thirumalai, Danda, & Krishnakumar, 2012). Another arti-
cle reports that curcumin and its derivative CDF regulate the expres- 4.8 | Curcumin analogues and their effect on the
sion of miRNAs such as miR-21, miR-22, miR-200b, mir-200c in removal of antitumor drug resistance
pancreatic cancer cells. Upregulation of miR-22 decreased the expres-
sion of the Sp1 transcription factor and ER 1, thus inhibited the cell Despite the widespread function of curcumin in inhibiting the growth
proliferation. Overexpression of miR-200b and 200c also induced the of cancer cells, its clinical use is problematic. The main problems of
reversal of epithelial-to-mesenchymal transition in gemcitabine- the use of curcumin are its poor solubility, low absorption, limited tis-
resistant pancreatic cancer cells (Simental-Mendía & Sahebkar, 2016). sue distribution, and rapid metabolism. One strategy to remove these
Therefore, curcumin, due to its importance in regulating the expression problems is to synthesize new curcumin analogues to increase its bio-
of miRNAs, can be considered as an important agent for cancer treat- availability while maintaining its ideal effectiveness. Modification of
ment as well as overcoming drug resistance. the curcumin structure enhanced its activity, penetration into the cell,
and increased the physiological and biological properties of curcumin.
So far, many high-efficacy curcumin analogs have been synthesized
4.7 | The effect of curcumin on CSCs (Mbese, Khwaza, & Aderibigbe, 2019; Murakami et al., 2017). One of
them is EF24, an excellent compound that has more bioactivity, anti-
Recently, numerous studies have shown that curcumin can inhibit tumor, and anti-inflammatory activity than curcumin. It inhibited cell
CSCs proliferation in various cancers. The inhibition can be due to cycle and induced apoptosis in cancer cells by inhibiting NF-κB,
impacts on CSC self-renewal pathways such as Wnt/b-catenin, notch, inhibiting HIF-1α, regulating ROS production, and regulating key
and sonic hedgehog. Several studies have demonstrated that cur- genes by miRNA. in vivo experiments have shown that EF24 sup-
cumin reduces catenin transcription in various cancer cells, leading to presses the growth of breast cancer cells with low toxicity in a mouse
decreased catenin levels, and inactivating the Wnt/b-catenin signaling xenograft model (He, Li, Hu, Sun, & Kong, 2018). Another report
pathway (Li & Zhang, 2014). The notch signaling pathway is also der- showed EF24 enhances the susceptibility of ovarian cancer resistant
egulated in CSCs. Curcumin treatment decreased notch-1 activity in cells to cisplatin via overexpression of p53 and p21 proteins in G2/M
esophageal squamous carcinoma cells through reducing γ-secretase checkpoint and consequently induces cellular arrest. It also induced
complex components expression. Curcumin also downregulated miR- apoptosis in cisplatin-resistant cells by activating PTEN phosphoryla-
21 and miR-34 as notch-1 specific microRNAs and significantly tion, inhibiting Akt and MDM2, and increasing p53 levels in the resis-
upregulated tumor suppressor let-7a miRNA. Therefore, curcumin tance cells (Selvendiran et al., 2007). Another analog curcumin is GO-
reduced cell proliferation and CSCs population in the surviving cells Y035, which showed more inhibitory effect on clonal cell growth cell
after treatment (Subramaniam et al., 2012). Downregulation and inac- line DLD-1 among the 2000 synthetic organic compound. Therefore,
tivation of notch-1 and NF-κB protein levels has also been reported in according to the GO-Y035 structure, more than 50 curcumin ana-
pancreatic and oral cancer cells (Li & Zhang, 2014; Subramaniam logues were synthesized. Studies of GO-Y030, GO-Y078, GO-Y168,
et al., 2012). Curcumin also can affect specific miRNAs in CSC to and GO-Y172 analogs on ABCG2 transporter have shown that these
remove the formation of the acquired EMT phenotype (Li, Kong, compounds can inhibit mitoxantrone and pheophorbide A efflux from
Wang, & Sarkar, 2010). For example, researchers have reported that the ABCG2 in resistance K562/BCRP cells. In addition, cytotoxicity
miR-200 reduces the formation of CSCs by downregulation of assays showed that GO-Y030 and GO-Y078 increase the efficacy of
notch-1 and EMT regulators. But, it is reduced in chemotherapy resis- SN-38 in these cells. Therefore, these analogs can remove the
tant cells which led to an increase in EMT phenotype. Studies have ABCG2-dependent MDR in the resistance cells (Murakami
shown curcumin and its analog CDF both increase the expression of et al., 2017). CDF is another analogue of curcumin. Its bioavailability is
miR-200b and miR-200c in gemcitabine-resistant pancreatic cancer high and various studies have shown a lower concentration is needed
cells and lead to the removal of EMT (Ali et al., 2010). The inhibitory compared to curcumin. The combination of CDF with gemcitabine
effects of curcumin on breast cancer stem cells (BCSCs) have also had a synergistic effect on pancreatic cancer cells. This combination
been studied. Curcumin deregulated the process of EMT in breast inhibited NF-κB activation and induced apoptosis in the cells. In addi-
cancer cells by reducing the proteins that play a key role in the forma- tion, CDF reduced the expression of miR-21 that is overexpressed in
tion of EMT such as Slug, AXL, twist1, N-cadherin, β-catenin, gemcitabine-resistant cells. Downregulation of miR-21 increased the
vimentin, and fibronectin (De, Beligala, Birkholz, & Geusz, 2019). The expression of PTEN and consequently induced cellular arrest
studies demonstrated the combination of curcumin with mitomycin C (Kwon, 2014). Another study showed that combination of CDF with
allows BCSCs to grow only the fourth generation. The size of the 5-fluorouracil and oxaliplatin (5-FU + Ox) had more growth-inhibitory
KEYVANI-GHAMSARI ET AL. 13

F I G U R E 9 Chemical
structures of curcumin (a) and its
novel synthetic analogs (b).
Reprinted with permission from
Mimeault and Batra (2011)
[Colour figure can be viewed at
wileyonlinelibrary.com]

in resistant colon cancer cells than curcumin. It reduced the expres- of curcumin is the use of delivery systems. A proper delivery system
sion of ABCG2 in the resistant colon cancer cells and strongly should increase the activity and toxicity of curcumin over free cur-
inhibited the growth of the CSCs. This combination inhibited activa- cumin, while not having side effects on the normal cells. Among many
tion of EGFR and IGF-1R signaling pathways, reduced activation of nano-based delivery groups, we will discuss some of them in this
β-catenin and NF-κB, downregulated the expression of c-Myc and section.
Cox-2, and increased proapoptotic protein Bax and caspase-3 activity. Liposomes are one of curcumin delivery systems. They consist of
Therefore, compared to curcumin, CDF induced a higher percentage two phospholipid layers and one aqueous nucleus therefore can carry
of apoptosis in chemoresistant colon cancer cells (Kanwar both hydrophobic and hydrophilic compounds. Liposomes increase
et al., 2011). Tripathi and Misra have designed a number of new cur- the stability of curcumin, its gastrointestinal absorption, and conse-
cumin analogues (compounds 1–6) that bind to P-gp at the surface of quently increase curcumin level in plasma (Yallapu, Jaggi, &
BCSCs and inhibited drug efflux process, thereby removing drug resis- Chauhan, 2013). Various types of polymers have been used to design
tance in these cells and inhibiting cancer cell growth. Among the nanoparticles for deliver curcumin. Synthetic polymers and natural
designed analogues, glucoside of curcumin was more likely to bind to polymers used in drug delivery systems must be biocompatible and
P-gp in the BCSCs (Tripathi & Misra, 2016). Dimethoxy-curcumin biodegradable. Due to these properties, synthetic polymers such as
(DMC) is a lipophilic derivative of curcumin. It has been recognized as PLGA (poly(D, L-lactic-co-glycolic acid)) is commonly used in the cur-
a potent anticancer agent in colon cancer. DMC-induced apoptosis in cumin delivery system (Tomeh, Hadianamrei, & Zhao, 2019). A num-
cloned cancer cells with less toxicity and side effects on normal cells ber of studies have shown that curcumin-loaded PLGA nanoparticles
than curcumin (Mbese et al., 2019). Therefore, as mentioned, the cur- (Cur-NPs) increase the oral bioavailability of curcumin. This formula-
cumin derivatives (Figure 9) overcome the disadvantages of curcumin tion increased water solubility, permeability, and inhibited P-gp
to enhance effectiveness of curcumin in cancer cell and remove drug mediated-efflux of curcumin. Another study reported curcumin-
resistance. loaded PLGA nanoparticles conjugated to P-gp (CUR-NPs-APgp) indi-
cated a higher affinity for drug-resistant cervical carcinoma cell line
KB-V1compared to curcumin and Cur-NPs. Given that P-gp is over-
4.9 | Curcumin delivery systems: Role of expressed in these cells and it is associated with aggressive behaviors
nanotechnology and MDR in KB-V1 cells, use of this formulation increased the uptake
of curcumin and enhanced its toxicity in these cells (Punfa, Yodkeeree,
In addition to the synthesis of curcumin derivatives, another strategy Pitchakarn, Ampasavate, & Limtrakul, 2012). Another study reported
that can overcome the low bioavailability and poor pharmacokinetic the effect of encapsulating curcumin and paclitaxel in flaxseed oil
14 KEYVANI-GHAMSARI ET AL.

containing nanoemulsion in ovarian cancer resistant cells. This formu- activation (Yan, Graham, & Lanza-Jacoby, 2013). High levels of flap
lation effectively delivers the drugs to the cells. NF-κB activity and P- endonuclease 1 (FEN1) expression has been observed in cisplatin
gp expression decreased in the resistant cells and apoptosis signifi- resistance in breast cancer cells. FEN1 is an enzyme involved in DNA
cantly increased in the presence of this combination (Ganta & replication and BER. Curcumin effectively reduced breast cancer cells
Amiji, 2009). Application of curcumin nanoparticle formulation has resistance to cisplatin through decreasing FEN1 expression (Zou
been shown to strongly inhibit cell growth and induce apoptosis in et al., 2018). Combination of curcumin with 5-Fluorouracil (5-FU) sig-
cervical cancer cells compared to free curcumin (Zaman et al., 2016). nificantly increased cell apoptosis in breast cancer cells compared to
Formulation of curcumin as curcumin/MPEG-PLA micelles increased 5-FU alone and chemosensitized cells to 5-FU. Repeated administra-
the toxicity of curcumin in a way that induced a higher percentage of tion of 5-FU increases the expression of thymidylate synthase
melanoma cells to apoptosis (Wang et al., 2017). Other researchers (TS) and consequently activates NF-κB, which results in resistance to
have found curcumin-loaded lipo-PEG PEI complexes (LPPC) increases 5-Fu in breast cancer cells. Research has shown that concomitant use
curcumin accumulation in B16F10 cells and enhances the toxicity of of curcumin and 5-Fu downregulates TS and inactivates NF-κB,
curcumin up to fivefold in curcumin-sensitive cells and up to 20-fold thereby improved the therapeutic effect of 5-FU (Tan &
in curcumin-resistant cells. Curcumin/LPPC inhibited cell cycle at Norhaizan, 2019; Vinod et al., 2013). Falah et al. studied the combina-
G2/M phase and increased the induction of apoptosis in the cells (Lin tion of metformin and curcumin against breast cancer in mice. They
et al., 2012). LDH has been used as drug delivery system due to their suggested the combination of metformin and curcumin showed the
good biocompatibility, low cytotoxicity, unique anionic exchange highest effect against cell proliferation in vitro and tumor growth
property, and pH controlled release property in acidic environments in vivo. The action of curcumin against angiogenesis was augmented
(especially cancer cells environment; Rives, Del Arco, & Martín, 2013). by its combination with metformin. Moreover, the combination
Recently Khorsandi et al. intercalated curcumin into LDH nanohybrid showed the highest effect on necrosis enhancement and apoptosis
to remove the drug resistance and enhance the effectiveness of the induction. It is also safe and did not exhibit toxicity side effects
curcumin in photodynamic therapy against breast cancer cells against liver and kidney functions (Falah, Talib, & Shbailat, 2017).
(Khorsandi et al., 2015, 2019). Silica-based nanoparticles also use as It has been seen that curcumin increases the sensitivity of the
deliver system for curcumin due to their resistant to changes in pH, colon cells to vincristine, cisplatin, and fluorouracil by inhibiting P-gp
temperature, microbial, and enzymatic attack. (Couleaud et al., 2010). and MDR (Lee, Joung, Cho, Son, & Lee, 2018). 5-FU, oxaliplatin, and
Mirzahosseinipour et al. showed that the antimicrobial and antibiofilm irinotecan are common drugs for the treatment of colone cancer that
activities of curcumin-silica nanoparticle-aPDT against P. aeruginosa generally, the efflux of these drugs from the cells by ABC transporters
and S. aureus was increased compare to alone curcumin reduces their effectiveness. Curcumin has been shown to suppress P-
(Mirzahosseinipour, Khorsandi, Hosseinzadeh, Ghazaeian, & gp (ABCB1), MRP1 (ABCC1), BCRP (ABCG2) and increase the toxicity
Shahidi, 2020). of these drugs in the colon cancer cells (Hu, Li, Gao, & Cho, 2016). In
addition, it has significantly removed 5-FU resistance in HCT-8 cell
line through downregulation of P-gp and HSP-27 and with reduction
4.10 | Combination of curcumin with anticancer in survivin and Bcl-2 expression induced apoptosis in these cells (Fan,
drugs Abulimiti, Zhang, Zhou, & Zhu, 2017). Another finding has shown pre-
treatment of HCT116R cells with curcumin effectively reduced the
Recently, significant consideration has been made on combination 5-fu resistance in resistant MMR deficient cells (Shakibaei
chemotherapy in cancer using plant derived-bioactive substances. In et al., 2014). Combination of camptothecin and curcumin-loaded cat-
this part, we will explain recent studies in the combination of cur- ionic polymeric nanoparticle increased intracellular drug concentra-
cumin with different anticancer drugs. tions and synergistic effects of the drugs in the colon-26 cells (Tan &
Meiyanto et al. has shown that combination of curcumin and its Norhaizan, 2019). Studies have shown that curcumin inhibits prolifer-
derivatives with doxorubicin enhance the toxicity of doxorubicin in ation of colon cancer by suppressing NF-κB activity and down-
resistant breast cancer cells through inhibition of HER2 and reduction regulation of COX −2. In addition, inhibiting NF-κB blocks the
of NF-κB activation (Meiyanto et al., 2014). Royt et al. reported that expression of MMP-9 and prevents invasion and metastasis of cancer
curcumin overcomes the drug resistance of cyclophosphamide and cells (Park, 2010). One of the reasons for the development of
paclitaxel in breast cancer cells via downregulation of telomerase, NF- irinotecan resistance is EMT in colon cancer. Research has shown that
κB, and HDAC (Wei, Pu, & Zhao, 2017). Curcumin enhances the effect curcumin significantly reduces irinotecan resistance and inhibits cell
of a HDAC inhibitor such as tricostatin A on cell cycle arrest in G0/G1 proliferation by increasing E-cadherin expression and decreasing
phase in breast cancer cell lines MDA-MB435eB and SkBr3. This com- vimentin and N-cadherin expression, which are responsible proteins
bination showed no change in p53 expression, increased p21, p27 and for the EMT process. Thus, curcumin increases the sensitivity of the
reduced cyclin D1 protein expression. Also, curcumin increased pro- colon cancer cells to irinotecan and induces apoptosis by removing
tein expression of phosphorylated JNK and induced cleavage of the EMT process (Zhang et al., 2018).
caspase 3 and PARP. These results suggested combination of cur- Curcumin could inhibited transcription of HPV 16 E6/E7,
cumin with tricostatin A induce p53 independent apoptosis via JNK upregulated p53, Rb, and PTPN13 expression in cervical cancer cells
KEYVANI-GHAMSARI ET AL. 15

(Mishra & Das, 2015). Other authors have also reported that and thereby activated APAF-1 and caspase-9, which increased apo-
coadministration of curcumin and paclitaxel has a synergistic effect on ptosis induction in cancer cells (Baharuddin et al., 2016). One of the
the treatment of cervical cancer. Curcumin increased susceptibility of reasons for cisplatin resistance is related to the level of copper trans-
cervical cancer cells to paclitaxel via downregulation of NF-κB and porter 1 (CTR1) at the cell surface, which plays a key role in platinum
Akt, increasing expression of p53, activating caspase 3, -7, -8, and -9, uptake. The amount of CTR1 is inversely related to the amount of
and release of cytochrome c (Bava et al., 2011; Dang, Yuan, Tian, Li, & copper in the cells. The researches illustrated curcumin can act as a
Liu, 2015; Wei et al., 2017). Curcumin by downregulation of HDACs, copper chelating agent, thus increasing the expression of CTR1 and
induced acetylation of p53 and overexpressed p53 in the cervical can- enhancing the absorption of cisplatin in the non-small-cell lung cancer
cer cell line SiHa and a drug-resistant clone SiHaR, lead to cell cycle (NSCLC), thereby increasing the sensitivity of tumor cells to cisplatin
arrest in phase G1-S. In addition, it reversed the drug resistance of in vivo and in vitro (Zhang et al., 2018). Another study has proved that
cervical cancer cells to cisplatin by suppression of MRP1 and P-gp1, curcumin enhances the toxicity of gefitinib in NSCLC by down-
therefore cells need a lower dose of drug (Roy & Mukherjee, 2014; regulating EGFR protein. Combination of curcumin with gefitinib also
Figure 10). inhibits Sp1, which reduces the expression of Sp1-dependent pro-
In vitro and in vivo studies have demonstrated that curcumin has teins, such as survivin, VEGF, and c-Met, in a manner that strongly
toxic effect on doxorubicin-resistant melanoma cells and induces apo- inhibits the growth of primary resistant NSCLC cells and can induce
ptosis in them (Odot et al., 2004). In A375 and C8161 cells, it arrested autophagy and apoptosis in the cells (Chen et al., 2019). in vivo and
cells at G2/M phase, and suppressed cell invasion via downregulation in vitro studies have shown curcumin inhibits angiogenesis by block-
PI3K/AKT/mTOR/P70S6K signaling pathway (ZHAO et al., 2016). ing PI3K/Akt/mTOR signaling pathway, inhibits hepatocyte growth
Another study has shown the combination of curcumin and tamoxifen factor-induced migration and induces apoptosis in lung cancer cells. In
at low doses induces apoptosis in the melanoma drug-resistant cells the experimental mouse model, curcumin increased the expression of
by increasing of ROS generation. In addition, it significantly induced E-cadherin and downregulated the expression of vimentin, CD34, and
autophagy in the cancer cells. Importantly, this combination does not VEGF and thereby inhibited EMT (Jiao et al., 2016).
affect noncancerous cells (Chatterjee & Pandey, 2011). Curcumin Peng et al. reported overexpression of P-gp in imatinib-resistant
decreased ethoposide resistance in melanoma cells by decreasing K562 cells (Peng, Tiwari, Wu, & Chen, 2012). Curcumin enhanced the
MPR1 and GST expression (Depeille, Cuq, Passagne, Evrard, & sensitivity of K562 cells to this drug by downregulation of P-gp. Also,
Vian, 2005). combination of curcumin and imatinib increased apoptosis induction
Studies have shown that combination of cisplatin with curcumin in the cells by reducing expression of NF-κB, surviving, HSP90, Bcl2
enhances the effectiveness of cisplatin in lung cancer cell lines. The genes and increasing caspase activity (Mukherjee, Sarkar, Mukherjee,
combined reduced cyclin D1 expression, increased p21 expression Biswas, & Roy, 2016; Peng et al., 2012).

F I G U R E 1 0 Co-treatment with curcumin and chemotherapy drugs. Curcumin increases chemosensitivity of anticancer drugs by changing
different signaling pathway. Reproduced with permission from Tan and Norhaizan (2019) [Colour figure can be viewed at wileyonlinelibrary.com]
16 KEYVANI-GHAMSARI ET AL.

5 | C U R CU M I N I N C LI NI CA L TR I A L S experienced hemolysis and three patients experienced significantly


reduced hemoglobin (Greil et al., 2018). Other researcher was con-
The first clinical use of curcumin carried out in 1937 against cholecys- ducted in Phase II trial of curcumin in the pancreatic cancer. For this
titis, but its clinical trial alone or in combination with other compounds purpose, 25 patients received 8 g/day of curcumin orally as their dis-
has been widely done for about a quarter of a century against various ease progressed with a break of 2 months. Analysis of curcumin phar-
cancers. Its use has been approved in many parts of the world, and as macology suggested low bioavailability of curcumin was improved.
mentioned, various types of delivery systems have been designed to The toxicity of the drug was not seen. Two patients responded to
increase its bioavailability (Shanmugam et al., 2015). Pharmacological treatment, and one patient was stable for more than 18 months and
effects of curcumin as oral formulation in a dose-finding design were another patient showed brief tumor response. The expression of NF-
explored in 15 patients who had advanced colorectal cancer resistant κB, COX-2, and pSTAT3 reduced in peripheral blood mononuclear
to chemotherapy. Patients received doses of 0.45–3.6 g daily for cells of patients (Dhillon et al., 2008). In another study, Phase I/II trial,
approximately 4 months. Curcumin levels and its metabolites were the effect of combination of curcumin with gemcitabine was evalu-
measured in plasma, urine, and feces. Also, the level of glutathione S- ated in gemcitabine-resistant pancreatic cancer patients. The
transferase activity and prostaglandin E2 (PGE2) production in blood 21 patients were given 8 g of curcumin orally every day plus a stan-
were measured as biomarkers of curcumin activity. Dose-limiting tox- dard dose of gemcitabine. Since no toxicity was seen in patients in
icity was not seen. The concentration of 10 nmol/L of curcumin and Phase I, the same drug concentration was selected for the Phase II
its glucuronide and sulfate metabolites were observed in plasma and study. Outcome was safe and well tolerated in all patients in this
in urine. The daily administration of 3.6 g reduced PGE2 production in study. Median survival time was 161 days. Further studies are needed
blood samples were collected 1 hr after consumption by 62 and 57% to complete the results on this satisfactory combination (Kanai
on Days 1 and 29, respectively. According to the results an oral dose et al., 2011). Thus clinical results have shown that curcumin alone or
of 3.6 g/day was proposed is suitable for the Phase II trial to prevent in combination with other drugs even up to the concentration of 8 g/
or treat outside the gastrointestinal tract (Sharma et al., 2004). day is safe, tolerable and without side effects in normal cells in various
Another study in a Phase I clinical trial showed that administration of cancers. However, its bioavailability is still a problem and the use of
3.6 mg curcumin three times daily for 10–30 days reduced serum curcumin analogues and its delivery systems are in the early stages.
tumor necrosis factor alpha (TNF-α) levels in patients with colon can- Nevertheless, curcumin has been introduced as a promising com-
cer. On the other hand, enhanced P53 and Bax expression in tumor pound in the treatment of cancer (Shanmugam et al., 2015).
tissues, thereby increased apoptosis in tumor cells (He et al., 2011).
Bayet-Robert et al. in a Phase I clinical trial in 14 patients with
advanced and metastatic breast cancer evaluated the combination of 6 | CONC LU SION
docetaxel 100 mg/m2 every 3 weeks during six cycles of chemother-
apy with curcumin 0.5–8 g/day for seven consecutive days per cycle. Despite significant advances in the production of new chemotherapy
A partial response was seen in some patients, but no progress was drugs, cancer treatment generally fails due to drug resistance. Drug
observed in any patients. The authors suggested that inhibition of resistance is caused by multiple mechanisms such as drug efflux, DNA
cancer progression can be due to the effect of curcumin on down- repair, drug inactivation or its target alteration, apoptosis breakdown,
regulation of P-gp that increased docetaxel absorption and removed and epigenetic changes (Housman et al., 2014; Klopfleisch
the resistance of cells to it. VEGF levels reduced significantly after et al., 2016). Nevertheless, combination therapy can be the best
three cycles of treatment. The results suggested that the best tolera- approach compared with the monotherapy to reduce the factors caus-
ble dose of curcumin can be 6 g/day in combination with a standard ing resistance (Mokhtari et al., 2017). As we described in this review,
dose of docetaxel for Phase II clinical trials. (Bayet-Robert curcumin has a significant effect on the cancer prevention, inhibiting
et al., 2010). Another study was conducted on theracurmin (a new cancer progression, induction of apoptosis, and by various mecha-
form of curcumin) in pancreatic cancer. The 16 patients took nisms removes the drug resistance and enhances efficacy of chemo-
theracurmin daily with standard gemcitabine concentration for therapy. Curcumin modulates signaling pathways of immune system,
6 months. The results provided safety and high bioavailability of cell cycle, apoptosis, metastasis, angiogenesis in the cells by regulating
theracurmin. The quality of life of patients also increased. No signifi- the expression of proteins such as oncogenes, tumor suppressor
cant changes in NF-κB activity and cytokine levels were observed genes, pro- and antiapoptotic proteins, NF-κB, Akt, VEGF, STAT3,
(Kanai et al., 2013). Greil et al. investigated the safety and pharmaco- MAPKs, COX-2, EGFR, HER-2, check points of the cell cycle, and
logical effect of liposomal curcumin on patients with metastatic can- MAPK (Rahmani, Al Zohairy, Aly, & Khan, 2014; Tomeh et al., 2019;
cers. Liposomal curcumin was given by intravenous injection to Wei et al., 2017). Numerous articles have reported curcumin-based
32 patients. Its increasing dose started from 100 mg/m2 over 8 hr to inhibition of ABC transporters, DNA repair proteins, DNMTs, HDACs,
300 mg/m2 over 6 hr. During injection curcumin concentration in activation of apoptotic genes, and regulating regulation of the expres-
plasma was constant but declined rapidly after injection. Drug toxicity sion of metabolic enzymes which overcomes the drug resistance and
was not reported in doses of 100–300 mg/m2 over 8 hr in 26 patients. increases chemosensitivity in various types of cancers both in vitro
Of the six patients who were given 300 mg/m2 over 6 hr, one patient and in vivo (Hsieh et al., 2014; Ismail, Othman, Abas, Lajis, &
KEYVANI-GHAMSARI ET AL. 17

Naidu, 2019; Limtrakul et al., 2004; Liu et al., 2009; Lu et al., 2009; Baker, E. K., & El-Osta, A. (2003). The rise of DNA methylation and the
Reuter et al., 2011). Despite the high efficacy of curcumin in the treat- importance of chromatin on multidrug resistance in cancer. Experimen-
tal Cell Research, 290, 177–194. https://doi.org/10.1016/S0014-4827
ment of cancer, its low toxicity and high safety, its main problem is
(03)00342-2
poor water solubility, low stability, and low bioavailability. To over- Bava, S. V., Sreekanth, C. N., Thulasidasan, A. K. T., Anto, N. P.,
come this problem strategies such as structural modifications and cur- Cheriyan, V. T., Puliyappadamba, V. T., … Anto, R. J. (2011). Akt is
cumin nanoformulations have been devised to increase the uptake upstream and MAPKs are downstream of NF-κB in paclitaxel-induced
survival signaling events, which are down-regulated by curcumin con-
and stability of curcumin (Khorsandi et al., 2015, 2019; Rai
tributing to their synergism. The International Journal of Biochemistry &
et al., 2015; Tan & Norhaizan, 2019; Tomeh et al., 2019). Cell Biology, 43, 331–341. https://doi.org/10.1016/j.biocel.2010.
Therefore, understanding the function and mechanism of cur- 09.011
cumin, and the most appropriate way to enhance its bioavailability Bayet-Robert, M., Kwiatkowski, F., Leheurteur, M., Gachon, F.,
Planchat, E., Abrial, C., … Chollet, P. (2010). Phase I dose escalation
provide insights into how to use this powerful natural agent to
trial of docetaxel plus curcumin in patients with advanced and meta-
enhance the effectiveness of antitumor drugs. static breast cancer. Cancer Biology & Therapy, 9, 14. https://doi.org/
10.4161/cbt.9.1.10392
CONF LICT OF IN TE RE ST Bell, D. W., Gore, I., Okimoto, R. A., Godin-Heymann, N., Sordella, R.,
Mulloy, R., … Haber, D. A. (2005). Inherited susceptibility to lung can-
The authors declare no conflict of interest.
cer may be associated with the T790M drug resistance mutation in
EGFR. Nature Genetics, 37, 1315–1316. https://doi.org/10.1038/
ORCID ng1671
Khatereh Khorsandi https://orcid.org/0000-0001-6882-5031 Bibi, Z. (2008). Nutrition & metabolism role of cytochrome P450 in drug
interactions. Nutrition & Metabolism (London), 5, 1–10. https://doi.org/
10.1186/1743-7075-5-27
RE FE R ENC E S
Bora-Tatar, G., Dayangaç-Erden, D., Demir, A. S., Dalkara, S., Yelekçi, K., &
Aggarwal, S., Takada, Y., Singh, S., Myers, J. N., & Aggarwal, B. B. (2004). Erdem-Yurter, H. (2009). Molecular modifications on carboxylic acid
Inhibition of growth and survival of human head and neck squamous derivatives as potent histone deacetylase inhibitors: Activity and dock-
cell carcinoma cells by curcumin via modulation of nuclear factor-κB ing studies. Bioorganic & Medicinal Chemistry, 17, 5219–5228. https://
signaling. International Journal of Cancer, 111, 679–692. https://doi. doi.org/10.1016/j.bmc.2009.05.042
org/10.1002/ijc.20333 Boyanapalli, S. S. S., & Kong, A. N. T. (2015). “Curcumin, the king of
Alfarouk, K. O., Stock, C. M., Taylor, S., Walsh, M., Muddathir, A. K., spices”: Epigenetic regulatory mechanisms in the prevention of cancer,
Verduzco, D., … Rauch, C. (2015). Resistance to cancer chemotherapy: neurological, and inflammatory diseases. Current Pharmacology Reports,
Failure in drug response from ADME to P-gp. Cancer Cell International, 1, 129–139. https://doi.org/10.1007/s40495-015-0018-x
15, 1–13. https://doi.org/10.1186/s12935-015-0221-1 Boyer, J., McLean, E. G., Aroori, S., Wilson, P., McCulla, A., Carey, P. D., …
Ali, S., Ahmad, A., Banerjee, S., Padhye, S., Dominiak, K., Schaffert, J. M., … Johnston, P. G. (2004). Characterization of p53 wild-type and null iso-
Sarkar, F. H. (2010). Gemcitabine sensitivity can be induced in pancre- genic colorectal cancer cell lines resistant to 5-fluorouracil, oxaliplatin,
atic cancer cells through modulation of miR-200 and miR-21 expres- and irinotecan. Clinical Cancer Research, 10, 2158–2167. https://doi.
sion by curcumin or its analogue CDF. Cancer Research, 70, org/10.1158/1078-0432.CCR-03-0362
3606–3617. https://doi.org/10.1158/0008-5472.CAN-09-4598 Bozic, I., Reiter, J. G., Allen, B., Antal, T., Chatterjee, K., Shah, P., …
Allocati, N., Masulli, M., Di Ilio, C., & Federici, L. (2018). Glutathione trans- Nowak, M. A. (2013). Evolutionary dynamics of cancer in response to
ferases: Substrates, inihibitors and pro-drugs in cancer and neurode- targeted combination therapy. eLife, 2013, 1–15. https://doi.org/10.
generative diseases. Oncogene, 7, 8. https://doi.org/10.1038/s41389- 7554/eLife.00747
017-0025-3 Chan, H.-K., & Ismail, S. (2014). Side effects of chemotherapy among can-
Ambudkar, S. V., Kimchi-Sarfaty, C., Sauna, Z. E., & Gottesman, M. M. cer patients in a Malaysian general hospital: Experiences, perceptions
(2003). P-glycoprotein: From genomics to mechanism. Oncogene, 22, and informational needs from clinical pharmacists. Asian Pacific Journal
7468–7485. https://doi.org/10.1038/sj.onc.1206948 of Cancer Prevention, 15, 5305–5309. https://doi.org/10.7314/apjcp.
Aslam, M. S., Naveed, S., Ahmed, A., Abbas, Z., Gull, I., & Athar, M. A. 2014.15.13.5305
(2014). Side effects of chemotherapy in cancer patients and evaluation Chang, G. (2003). Multidrug resistance ABC transporters. FEBS Letters,
of patients opinion about starvation based differential chemotherapy. 555, 102–105. https://doi.org/10.1016/S0014-5793(03)01085-8
Journal of Cancer Therapy, 05, 817–822. https://doi.org/10.4236/jct. Chatterjee, S. J., & Pandey, S. (2011). Chemo-resistant melanoma sensi-
2014.58089 tized by tamoxifen to low dose curcumin treatment through induction
Aung, T., Qu, Z., Kortschak, R., & Adelson, D. (2017). Understanding the of apoptosis and autophagy. Cancer Biology & Therapy, 11, 216–228.
effectiveness of natural compound mixtures in Cancer through their https://doi.org/10.4161/cbt.11.2.13798
molecular mode of action. International Journal of Molecular Sciences, Chearwae, W., Shukla, S., Limtrakul, P., & Ambudkar, S. V. (2006). Modula-
18, 656. https://doi.org/10.3390/ijms18030656 tion of the function of the multidrug resistance-linked ATP-binding
Baharuddin, P., Satar, N., Fakiruddin, K. S., Zakaria, N., Lim, M. N., cassette transporter ABCG2 by the cancer chemopreventive agent
Yusoff, N. M., … Yahaya, B. H. (2016). Curcumin improves the efficacy curcumin. Molecular Cancer Therapeutics, 5, 1995–2006. https://doi.
of cisplatin by targeting cancer stem-like cells through p21 and cyclin org/10.1158/1535-7163.MCT-06-0087
D1-mediated tumour cell inhibition in non-small cell lung cancer cell Chen, P., Huang, H. P., Wang, Y., Jin, J., Long, W. G., Chen, K., … Li, J.
lines. Oncology Reports, 35, 13–25. https://doi.org/10.3892/or.2015. (2019). Curcumin overcome primary gefitinib resistance in non-small-
4371 cell lung cancer cells through inducing autophagy-related cell death.
Baig, S., Seevasant, I., Mohamad, J., Mukheem, A., Huri, H. Z., & Journal of Experimental & Clinical Cancer Research, 38, 1–17. https://
Kamarul, T. (2016). Potential of apoptotic pathway-targeted cancer doi.org/10.1186/s13046-019-1234-8
therapeutic research: Where do we stand? Cell Death & Disease, 7, Conseil, G., Baubichon-Cortay, H., Dayan, G., Jault, J.-M., Barron, D., & Di
e2058. https://doi.org/10.1038/cddis.2015.275 Pietro, A. (2002). Flavonoids: A class of modulators with bifunctional
18 KEYVANI-GHAMSARI ET AL.

interactions at vicinal ATP- and steroid-binding sites on mouse P-gly- Fardi, M., Solali, S., & Farshdousti Hagh, M. (2018). Epigenetic mechanisms
coprotein. Proceedings of the National Academy of Sciences of the as a new approach in cancer treatment: An updated review. Genes &
United States of America, 95, 9831–9836. https://doi.org/10.1073/ Diseases, 5, 304–311. https://doi.org/10.1016/j.gendis.2018.06.003
pnas.95.17.9831 Fulda, S., & Debatin, K.-M. (2006). Extrinsic versus intrinsic apoptosis path-
Couleaud, P., Morosini, V., Frochot, C., Richeter, S., Raehm, L., & ways in anticancer chemotherapy. Oncogene, 25, 4798–4811. https://
Durand, J.-O. (2010). Silica-based nanoparticles for photodynamic doi.org/10.1038/sj.onc.1209608
therapy applications. Nanoscale, 2, 1083–1095. https://doi.org/10. Ganta, S., & Amiji, M. (2009). Coadministration of paclitaxel and curcumin
1039/c0nr00096e in nanoemulsion formulations to overcome multidrug resistance in
Criscitiello, C., Fumagalli, D., Saini, K. S., & Loi, S. (2011). Tamoxifen in tumor cells. Molecular Pharmaceutics, 6, 928–939. https://doi.org/10.
early-stage estrogen receptor positive breast cancer: Overview of clin- 1021/mp800240j
ical use and molecular biomarkers for patient selection. Oncotargets Garc, M., Elisabet, P., & Redondo, M. 2018. Bcl-2 inhibition to overcome
and Therapy, 4, 1–11. https://doi.org/10.2147/OTT.S10155 resistance to chemo- and immunotherapy. doi:https://doi.org/10.
Dang, Y. P., Yuan, X. Y., Tian, R., Li, D. G., & Liu, W. (2015). Curcumin 3390/ijms19123950
improves the paclitaxel induced apoptosis of HPV positive human cer- García-Becerra, R., Santos, N., Díaz, L., & Camacho, J. (2013). Mechanisms
vical cancer cells via the NF κB p53 caspase 3 pathway. Experimental of resistance to endocrine therapy in breast cancer: Focus on signaling
and Therapeutic Medicine, 9, 1470–1476. https://doi.org/10.3892/ pathways, miRNAs and genetically based resistance. International Jour-
etm.2015.2240 nal of Molecular Sciences, 14, 108–145. https://doi.org/10.3390/
Danielson, P. B. (2002). The cytochrome P450 superfamily: Biochemistry, ijms14010108
evolution and drug metabolism in humans. Current Drug Metabolism, 3, Geisler, S., Børresen-Dale, A. L., Johnsen, H., Aas, T., Geisler, J.,
561–597. https://doi.org/10.2174/1389200023337054 Akslen, L. A., … Lønning, P. E. (2003). TP53 gene mutations predict the
De, A., Beligala, D. H., Birkholz, T. M., & Geusz, M. E. (2019). Anticancer response to neoadjuvant treatment with 5-fluorouracil and mitomycin
properties of curcumin and interactions with the circadian timing sys- in locally advanced breast cancer. Clinical Cancer Research, 9,
tem. Integrative Cancer Therapies, 18, 1–10. https://doi.org/10.1177/ 5582–5588.
1534735419889154 Ghasabi, M., Mansoori, B., Mohammadi, A., Duijf, P. H. G., Shomali, N.,
de Vries, E. G. E. (2006). Tumor necrosis factor-related apoptosis-inducing Shirafkan, N., … Baradaran, B. (2019). MicroRNAs in cancer drug resis-
ligand pathway and its therapeutic implications. Clinical Cancer tance: Basic evidence and clinical applications. Journal of Cellular Physi-
Research, 12, 2390–2393. https://doi.org/10.1158/1078-0432.CCR- ology, 234, 2152–2168. https://doi.org/10.1002/jcp.26810
06-0352 Goel, A., & Aggarwal, B. B. (2010). Curcumin, the Golden spice from Indian
Depeille, P., Cuq, P., Passagne, I., Evrard, A., & Vian, L. (2005). Combined saffron, is a chemosensitizer and radiosensitizer for tumors and
effects of GSTP1 and MRP1 in melanoma drug resistance. British Jour- chemoprotector and radioprotector for normal organs. Nutrition and Can-
nal of Cancer, 93(2), 216–223. https://doi.org/10.1038/sj.bjc.6602681 cer, 62, 919–930. https://doi.org/10.1080/01635581.2010.509835
Dhillon, N., Aggarwal, B. B., Newman, R. A., Wolff, R. A., Greil, R., Greil-Ressler, S., Weiss, L., Schönlieb, C., Magnes, T., Radl, B., …
Kunnumakkara, A. B., Abbruzzese, J. L., … Kurzrock, R. (2008). Phase II Sordillo, P. P. (2018). A phase 1 dose-escalation study on the safety,
trial of curcumin in patients with advanced pancreatic cancer. Clinical tolerability and activity of liposomal curcumin (Lipocurc™) in patients
Cancer Research, 14, 4491–4499. https://doi.org/10.1158/1078- with locally advanced or metastatic cancer. Cancer Chemotherapy and
0432.CCR-08-0024 Pharmacology, 82, 695–706. https://doi.org/10.1007/s00280-018-
Duvoix, A., Morceau, F., Delhalle, S., Schmitz, M., Schnekenburger, M., 3654-0
Galteau, M. M., … Diederich, M. (2003). Induction of apoptosis by cur- Hamed, A. R., Abdel-Azim, N. S., Shams, K. A., & Hammouda, F. M. (2019).
cumin: Mediation by glutathione S-transferase P1-1 inhibition. Bio- Targeting multidrug resistance in cancer by natural chemosensitizers.
chemical Pharmacology, 66, 1475–1483. https://doi.org/10.1016/ Bulletin of the National Research Centre, 43, 8. https://doi.org/10.
S0006-2952(03)00501-X 1186/s42269-019-0043-8
Eid, S. Y., El-Readi, M. Z., Fatani, S. H., Mohamed Nour Eldin, E. E., & Hassan, F., Rehman, M. S., Khan, M. S., Ali, M. A., Javed, A., Nawaz, A., &
Wink, M. (2015). Natural products modulate the multifactorial Yang, C. (2019). Curcumin as an alternative epigenetic modulator:
multidrug resistance of cancer. Journal of Pharmacy and Pharmacology, Mechanism of action and potential effects. Frontiers in Genetics, 10,
06, 146–176. https://doi.org/10.4236/pp.2015.63017 514. https://doi.org/10.3389/fgene.2019.00514
Ekor, M. (2014). The growing use of herbal medicines: Issues relating to He, Y., Li, W., Hu, G., Sun, H., & Kong, Q. (2018). Bioactivities of EF24, a
adverse reactions and challenges in monitoring safety. Frontiers in Neu- novel curcumin analog: A review. Frontiers in Oncology, 8, 614. https://
rology, 4, 1–10. https://doi.org/10.3389/fphar.2013.00177 doi.org/10.3389/fonc.2018.00614
Escárcega, R. O., Fuentes-Alexandro, S., García-Carrasco, M., Gatica, A., & He, Z. Y., Shi, C. B., Wen, H., Li, F. L., Wang, B. L., & Wang, J. (2011).
Zamora, A. (2007). The transcription factor nuclear factor-kappa B and Upregulation of p53 expression in patients with colorectal cancer by
cancer. Clinical Oncology, 19, 154–161. https://doi.org/10.1016/j.clon. administration of curcumin. Cancer Investigation, 29, 208–213. https://
2006.11.013 doi.org/10.3109/07357907.2010.550592
Fabian, M. R., Sonenberg, N., & Filipowicz, W. (2010). Regulation of mRNA Hervouet, E., Cartron, P., Jouvenot, M., & Delage-mourroux, R. (2013). Epi-
translation and stability by microRNAs. Annual Review of Biochemistry, genetic regulation of estrogen signaling in breast cancer. Epigenetics,
79, 351–379. https://doi.org/10.1146/annurev-biochem-060308- 2013, 237–245.
103103 Holohan, C., Van Schaeybroeck, S., Longley, D. B., & Johnston, P. G.
Falah, R. R., Talib, W. H., & Shbailat, S. J. (2017). Combination of metfor- (2013). Cancer drug resistance: An evolving paradigm. Nature Reviews.
min and curcumin targets breast cancer in mice by angiogenesis inhibi- Cancer, 13, 714–726. https://doi.org/10.1038/nrc3599
tion, immune system modulation and induction of p53 independent Housman, G., Byler, S., Heerboth, S., Lapinska, K., Longacre, M., Snyder, N., &
apoptosis. Therapeutic Advances in Medical Oncology, 9, 235–252. Sarkar, S. (2014). Drug resistance in cancer: An overview. Cancers (Basel).,
https://doi.org/10.1177/1758834016687482 6, 1769–1792. https://doi.org/10.3390/cancers6031769
Fan, Y. X., Abulimiti, P., Zhang, H. L., Zhou, Y. K., & Zhu, L. (2017). Mecha- Hsieh, Y. W., Huang, C. Y., Yang, S. Y., Peng, Y. H., Yu, C. P.,
nism of reversal of multidrug resistance by curcumin in human colorec- Chao, P. D. L., & Hou, Y. C. (2014). Oral intake of curcumin markedly
tal cancer cell line HCT-8/5-FU. Genetics and Molecular Research, 16, activated CYP 3A4: in vivo and ex-vivo studies. Scientific Reports, 4,
1–13. https://doi.org/10.4238/gmr16029414 1–7. https://doi.org/10.1038/srep06587
KEYVANI-GHAMSARI ET AL. 19

Hu, Q., & Baeg, G. H. (2017). Role of epigenome in tumorigenesis and drug Khorsandi, K., Hosseinzadeh, R., & Shahidi, F. K. (2019). Photodynamic
resistance. Food and Chemical Toxicology, 109, 663–668. https://doi. treatment with anionic nanoclays containing curcumin on human
org/10.1016/j.fct.2017.07.022 triple-negative breast cancer cells: Cellular and biochemical studies.
Hu, T., Li, Z., Gao, C. Y., & Cho, C. H. (2016). Mechanisms of drug resis- Journal of Cellular Biochemistry, 120, 4998–5009. https://doi.org/10.
tance in colon cancer and its therapeutic strategies. World Journal of 1002/jcb.27775
Gastroenterology, 22, 6876–6889. https://doi.org/10.3748/wjg.v22. Kitagawa, S., Nabekura, T., & Kamiyama, S. (2004). Inhibition of P-
i30.6876 glycoprotein function by tea catechins in KB-C2 cells. The Journal of
Huang, C.-Y., Ju, D.-T., Chang, C.-F., Muralidhar Reddy, P., & Pharmacy and Pharmacology, 56, 1001–1005. https://doi.org/10.
Velmurugan, B. K. (2017). A review on the effects of current chemo- 1211/0022357044003
therapy drugs and natural agents in treating non-small cell lung cancer. Klopfleisch, R., Kohn, B., & Gruber, A. D. (2016). Mechanisms of tumour
Biomedicine, 7, 23. https://doi.org/10.1051/bmdcn/2017070423 resistance against chemotherapeutic agents in veterinary oncology.
Iqbal, J., Abbasi, B. A., Mahmood, T., Kanwal, S., Ali, B., Shah, S. A., & Veterinary Journal, 207, 63–72. https://doi.org/10.1016/j.tvjl.2015.
Khalil, A. T. (2017). Plant-derived anticancer agents: A green antican- 06.015
cer approach. Asian Pacific Journal of Tropical Biomedicine, 7, Kovalchuk, O., Filkowski, J., Meservy, J., Ilnytskyy, Y., Tryndyak, V. P.,
1129–1150. https://doi.org/10.1016/j.apjtb.2017.10.016 Chekhun, V. F., & Pogribny, I. P. (2008). Involvement of microRNA-
Ismail, N. I., Othman, I., Abas, F., Lajis, N. H., & Naidu, R. (2019). Mecha- 451 in resistance of the MCF-7 breast cancer cells to chemotherapeu-
nism of apoptosis induced by curcumin in colorectal cancer. Interna- tic drug doxorubicin. Molecular Cancer Therapeutics, 7, 2152–2159.
tional Journal of Molecular Sciences, 20, 2454. https://doi.org/10. https://doi.org/10.1158/1535-7163.MCT-08-0021
3390/ijms20102454 Krausz, A. E., Adler, B. L., Cabral, V., Navati, M., Doerner, J.,
Jacob, A., Wu, R., Zhou, M., & Wang, P. (2007). Mechanism of the anti- Charafeddine, R. A., … Friedman, A. J. (2015). Curcumin-encapsulated
inflammatory effect of curcumin: PPAR-γ activation. PPAR Research, nanoparticles as innovative antimicrobial and wound healing agent.
2007, 1–5. https://doi.org/10.1155/2007/89369 Nanomedicine Nanotechnology. Biologie et Médecine, 11, 195–206.
Jalkanen, A. L., Coleman, S. J., & Wilusz, J. (2014). Determinants and impli- https://doi.org/10.1016/j.nano.2014.09.004
cations of mRNA poly(A) tail size—Does this protein make my tail look Kumar, S., Kushwaha, P. P., & Gupta, S. (2019). Emerging targets in cancer
big? Seminars in Cell & Developmental Biology, 34, 24–32. https://doi. drug resistance. Cancer Drug Resistance. 2, 161–177. https://doi.org/
org/10.1016/j.semcdb.2014.05.018 10.20517/cdr.2018.27
Ji, Z. (2010). Targeting DNA damage and repair by curcumin. Breast cancer: Kwon, Y. (2014). Curcumin as a cancer chemotherapy sensitizing agent.
Basic and Clinical Research, 4, 3–5. Journal of Korean Society for Applied Biological Chemistry, 57, 273–280.
Jiao, D., Wang, J., Lu, W., Tang, X., Chen, J., Mou, H., & Chen, Q. Y. (2016). https://doi.org/10.1007/s13765-014-4077-1
Curcumin inhibited HGF-induced EMT and angiogenesis through regu- Lee, G., Joung, J. Y., Cho, J. H., Son, C. G., & Lee, N. (2018). Overcoming P-
lating c-Met dependent PI3K/Akt/mTOR signaling pathways in lung glycoprotein-mediated multidrug resistance in colorectal cancer:
cancer. Molecular Therapy: Oncolytics, 3, 16018. https://doi.org/10. Potential reversal agents among herbal medicines. Alternative Medicine,
1038/mto.2016.18 2018, 9. https://doi.org/10.1155/2018/3412074
Jubierre, L., Jiménez, C., Rovira, E., Soriano, A., Sábado, C., Gros, L., … Lee, S. J., Krauthauser, C., Maduskuie, V., Fawcett, P. T., Olson, J. M., &
Segura, M. F. (2018). Targeting of epigenetic regulators in neuroblas- Rajasekaran, S. A. (2011). Curcumin-induced HDAC inhibition and
toma. Experimental & Molecular Medicine, 50, 51. https://doi.org/10. attenuation of medulloblastoma growth in vitro and in vivo. BMC Can-
1038/s12276-018-0077-2 cer, 11, 144. https://doi.org/10.1186/1471-2407-11-144
Kanai, M., Otsuka, Y., Otsuka, K., Sato, M., Nishimura, T., Mori, Y., … Lehmann, A. R. (2005). Replication of damaged DNA by translesion syn-
Shibata, H. (2013). A phase i study investigating the safety and phar- thesis in human cells. FEBS Letters, 579, 873–876. https://doi.org/10.
macokinetics of highly bioavailable curcumin (Theracurmin®) in cancer 1016/j.febslet.2004.11.029
patients. Cancer Chemotherapy and Pharmacology, 71, 1521–1530. Li, Y., Kong, D., Wang, Z., & Sarkar, F. H. (2010). Regulation of microRNAs
https://doi.org/10.1007/s00280-013-2151-8 by natural agents: An emerging field in chemoprevention and chemo-
Kanai, M., Yoshimura, K., Asada, M., Imaizumi, A., Suzuki, C., therapy research. Pharmaceutical Research, 27, 1027–1041. https://
Matsumoto, S., … Aggarwal, B. B. (2011). A phase I/II study of doi.org/10.1007/s11095-010-0105-y
gemcitabine-based chemotherapy plus curcumin for patients with Li, Y., & Zhang, T. (2014). Targeting cancer stem cells by curcumin and clin-
gemcitabine-resistant pancreatic cancer. Cancer Chemotherapy and ical applications. Cancer Letters, 346, 197–205. https://doi.org/10.
Pharmacology, 68, 157–164. https://doi.org/10.1007/s00280-010- 1016/j.canlet.2014.01.012
1470-2 Likhite, V. S., Stossi, F., Kim, K., Katzenellenbogen, B. S., &
Kanwal, R., & Gupta, S. (2012). Epigenetic modifications in cancer. Clinical Katzenellenbogen, J. A. (2006). Kinase-specific phosphorylation of the
Genetics, 81, 303–311. https://doi.org/10.1111/j.1399-0004.2011. estrogen receptor changes receptor interactions with ligand,
01809.x deoxyribonucleic acid, and coregulators associated with alterations in
Kanwar, S. S., Yu, Y., Nautiyal, J., Patel, B. B., Padhye, S., Sarkar, F. H., & estrogen and tamoxifen activity. Molecular Endocrinology, 20,
Majumdar, A. P. N. (2011). Difluorinated-curcumin (CDF): A novel cur- 3120–3132. https://doi.org/10.1210/me.2006-0068
cumin analog is a potent inhibitor of colon cancer stem-like cells. Phar- Limtrakul, P., Anuchapreeda, S., & Buddhasukh, D. (2004). Modulation of
maceutical Research, 28, 827–838. https://doi.org/10.1007/s11095- human multidrug-resistance MDR-1 gene by natural. BMC Cancer,
010-0336-y 6, 1–6.
Khazaei Koohpar, Z., Entezari, M., Movafagh, A., & Hashemi, M. (2015). Lin, Y. L., Liu, Y. K., Tsai, N. M., Hsieh, J. H., Chen, C. H., Lin, C. M., &
Anticancer activity of curcumin on human breast adenocarcinoma: Liao, K. W. (2012). A Lipo-PEG-PEI complex for encapsulating cur-
Role of Mcl-1 gene. Iranian Journal of Cancer Prevention, 8, e2331. cumin that enhances its antitumor effects on curcumin-sensitive and
https://doi.org/10.17795/ijcp2331 curcumin-resistance cells. Nanomedicine Nanotechnology. Biologie et
Khorsandi, K., Hosseinzadeh, R., & Fateh, M. (2015). Curcumin intercalated Médecine, 8, 318–327. https://doi.org/10.1016/j.nano.2011.06.011
layered double hydroxide nanohybrid as a potential drug delivery sys- Liu, F. S. (2009). Mechanisms of chemotherapeutic drug resistance in can-
tem for effective photodynamic therapy in human breast cancer cells. cer therapy—A quick review. Taiwanese Journal of Obstetrics & Gyne-
RSC Advances, 5, 93987–93994. https://doi.org/10.1039/ cology, 48, 239–244. https://doi.org/10.1016/S1028-4559(09)
C5RA15888E 60296-5
20 KEYVANI-GHAMSARI ET AL.

Liu, H.-T., & Ho, Y.-S. (2018). Anticancer effect of curcumin on breast can- Mirzaei, H., Masoudifar, A., Sahebkar, A., Zare, N., Nahand, J. S.,
cer and stem cells. Food Science and Human Wellness, 7, 134–137. Rashidi, B., … Jaafari, M. R. (2017). MicroRNA: A novel target of cur-
https://doi.org/10.1016/j.fshw.2018.06.001 cumin in cancer therapy. Journal of Cellular Physiology, 233,
Liu, H. L., Chen, Y., Cui, G. H., & Zhou, J. F. (2005). Curcumin, a potent 3004–3015. https://doi.org/10.1002/jcp.26055
anti-tumor reagent, is a novel histone deacetylase inhibitor regulating Mirzaei, H., Momeni, F., Saadatpour, L., Sahebkar, A., Goodarzi, M.,
B-NHL cell line Raji proliferation. Acta Pharmacologica Sinica, 26, Masoudifar, A., … Jaafari, M. R. (2018). MicroRNA: Relevance to stroke
603–609. https://doi.org/10.1111/j.1745-7254.2005.00081.x diagnosis, prognosis, and therapy. Journal of Cellular Physiology, 233,
Liu, Z., Xie, Z., Jones, W., Pavlovicz, R. E., Liu, S., Yu, J., … Chan, K. K. 856–865. https://doi.org/10.1002/jcp.25787
(2009). Curcumin is a potent DNA hypomethylation agent. Bioorganic & Mirzahosseinipour, M., Khorsandi, K., Hosseinzadeh, R., Ghazaeian, M., &
Medicinal Chemistry Letters, 19, 706–709. https://doi.org/10.1016/j. Shahidi, F. K. (2020). Antimicrobial photodynamic and wound healing
bmcl.2008.12.041 activity of curcumin encapsulated in silica nanoparticles. Pho-
Lu, H. F., Yang, J. S., Lai, K. C., Hsu, S. C., Hsueh, S. C., Chen, Y. L., … todiagnosis and Photodynamic Therapy, 29, 101639. https://doi.org/10.
Chung, J. G. (2009). Curcumin-induced DNA damage and inhibited 1016/j.pdpdt.2019.101639
DNA repair genes expressions in mouse-rat hybrid retina neuroblas- Mishra, A., & Das, B. C. (2015). Curcumin as an anti-human papillomavirus
toma cells ganglion cells (n18). Neurochemical Research, 34, and anti-cancer compound. Future Oncology, 11, 2487–2490. https://
1491–1497. https://doi.org/10.1007/s11064-009-9936-5 doi.org/10.2217/fon.15.166
Ma, J., Dong, C., & Ji, C. (2010). MicroRNA and drug resistance. Cancer Misra, R., & Sahoo, S. K. (2011). Coformulation of doxorubicin and cur-
Gene Therapy, 17, 523–531. https://doi.org/10.1038/cgt.2010.18 cumin in poly(d,l-lactide-co-glycolide) nanoparticles suppresses the
Malhotra, V., & Perry, M. C. (2003). Classical chemotherapy: Mechanisms, development of multidrug resistance in K562 cells. Molecular Pharma-
toxicities and the therapeutc window. Cancer Biology & Therapy, 2, ceutics, 8, 852–866. https://doi.org/10.1021/mp100455h
1–3. https://doi.org/10.4161/cbt.199 Mokhtari, R. B., Homayouni, T. S., Baluch, N., Morgatskaya, E., Kumar, S.,
Mansoori, B., Mohammadi, A., Davudian, S., Shirjang, S., & Baradaran, B. Das, B., & Yeger, H. (2017). Combination therapy in combating cancer.
(2017). The different mechanisms of cancer drug resistance: A brief Oncotarget, 8, 38022–38043. https://doi.org/10.18632/oncotarget.
review. Advanced Pharmaceutical Bulletin, 7, 339–348. https://doi.org/ 16723
10.15171/apb.2017.041 Mondal, S., Bhattacharya, K., & Mandal, C. (2018). Nutritional stress repro-
Mapoung, S., Pitchakarn, P., Yodkeeree, S., Ovatlarnporn, C., Sakorn, N., & grams dedifferention in glioblastoma multiforme driven by
Limtrakul, P. (2016). Chemosensitizing effects of synthetic curcumin ana- PTEN/Wnt/hedgehog axis: A stochastic model of cancer stem cells. Cell
logs on human multi-drug resistance leukemic cells. Chemico-Biological Death Discovery, 4, 110. https://doi.org/10.1038/s41420-018-0126-6
Interactions, 244, 140–148. https://doi.org/10.1016/j.cbi.2015.12.001 Mori, Y., Tatematsu, K., Koide, A., Sugie, S., Tanaka, T., & Mori, H. (2006).
Mathew, N., Kalyanasundaram, M., & Balaraman, K. (2006). Glutathione S- Modification by curcumin of mutagenic activation of carcinogenic N-
transferase (GST) inhibitors. Expert Opinion on Therapeutic Patents, 16 nitrosamines by extrahepatic cytochromes P-450 2B1 and 2E1 in rats.
(4), 431–444. Cancer Science, 97, 896–904. https://doi.org/10.1111/j.1349-7006.
Mbese, Z., Khwaza, V., & Aderibigbe, B. A. (2019). Curcumin and its deriv- 2006.00261.x
atives as potential therapeutic agents in prostate, colon and breast Mortezaee, K., Najafi, M., Farhood, B., Ahmadi, A., Shabeeb, D., &
cancers. Molecules, 24, 4386. https://doi.org/10.3390/molecules Musa, A. E. (2019). NF-κB targeting for overcoming tumor resistance
24234386 and normal tissues toxicity. Journal of Cellular Physiology, 234,
Meiyanto, E., Putri, D. D. P., Susidarti, R. A., Murwanti, R., Sardjiman, S., 17187–17204. https://doi.org/10.1002/jcp.28504
Fitriasari, A., … Kawaichi, M. (2014). Curcumin and its analogues Mudduluru, G., George-William, J. N., Muppala, S., Asangani, I. A.,
(PGV-0 and PGV-1) enhance sensitivity of resistant MCF-7 cells to Kumarswamy, R., Nelson, L. D., & Allgayer, H. (2011). Curcumin regu-
doxorubicin through inhibition of HER2 and NF-kB activation. Asian lates miR-21 expression and inhibits invasion and metastasis in colo-
Pacific Journal of Cancer Prevention, 15, 179–184. https://doi.org/10. rectal cancer. Bioscience Reports, 31, 185–197. https://doi.org/10.
7314/apjcp.2014.15.1.179 1042/BSR20100065
Meng, W., Ze-Fa, L., Hua, T., & Bao-An, C. (2018). Application of alkaloids Mukherjee, A., Sarkar, R., Mukherjee, S., Biswas, J., & Roy, M. (2016). Cur-
in reversing multidrug resistance in human cancers. Chinese Journal of cumin boosts up the efficacy of imatinib mesylate in chronic
Natural Medicines, 16, 561–571. https://doi.org/10.1016/S1875-5364 myelogenic leukemia cell line K-562 by modulation of various markers.
(18)30093-1 International Journal of Current Microbiology and Applied Sciences, 5,
Michael, M., & Doherty, M. M. (2005). Tumoral drug metabolism: Over- 240–255. https://doi.org/10.20546/ijcmas.2016.512.026
view and its implications for cancer therapy. Journal of Clinical Oncol- Müller, M. R., Thomale, J., Rajewsky, M. F., & Seeber, S. (1998). Drug resis-
ogy, 23, 205–229. https://doi.org/10.1200/JCO.2005.02.120 tance and DNA repair in leukaemia. Multidrug Resistance Cancer, 2,
Miller, T. E., Ghoshal, K., Ramaswamy, B., Roy, S., Datta, J., Shapiro, C. L., 175–185. https://doi.org/10.1007/978-94-017-2374-9_11
… Majumder, S. (2008). MicroRNA-221/222 confers tamoxifen resis- Murakami, M., Ohnuma, S., Fukuda, M., Chufan, E. E., Kudoh, K.,
tance in breast cancer by targeting p27Kip1. The Journal of Biological Kanehara, K., … Unno, M. (2017). Synthetic analogs of curcumin modu-
Chemistry, 283, 29897–29903. https://doi.org/10.1074/jbc. late the function of multidrug resistance–linked ATP-binding cassette
M804612200 transporter ABCG2. Drug Metabolism and Disposition, 45, 1166–1177.
Millimouno, F. M., Dong, J., Yang, L., Li, J., & Li, X. (2014). Targeting apo- https://doi.org/10.1124/dmd.117.076000
ptosis pathways in cancer and perspectives with natural compounds Nabekura, T. (2010). Overcoming multidrug resistance in human cancer
from mother nature. Cancer Prevention Research, 7, 1081–1107. cells by natural compounds. Toxins (Basel)., 2, 1207–1224. https://doi.
https://doi.org/10.1158/1940-6207.CAPR-14-0136 org/10.3390/toxins2061207
Mimeault, M., & Batra, S. K. (2011). Potential applications of curcumin and Nabekura, T., Yamaki, T., Ueno, K., & Kitagawa, S. (2008). Inhibition of P-
its novel synthetic analogs and nanotechnology-based formulations in glycoprotein and multidrug resistance protein 1 by dietary phytochem-
cancer prevention and therapy. Chinese Medicine, 6, 31. https://doi. icals. Cancer Chemotherapy and Pharmacology, 62, 867–873. https://
org/10.1186/1749-8546-6-31 doi.org/10.1007/s00280-007-0676-4
Miremadi, A., Oestergaard, M. Z., Pharoah, P. D. P., & Caldas, C. (2007). Najafi, M., Mortezaee, K., & Ahadi, R. (2019). Cancer stem cell (a)symme-
Cancer genetics of epigenetic genes. Human Molecular Genetics, 16, try & plasticity: Tumorigenesis and therapy relevance. Life Sciences,
R28–R49. https://doi.org/10.1093/hmg/ddm021 231, 116520. https://doi.org/10.1016/j.lfs.2019.05.076
KEYVANI-GHAMSARI ET AL. 21

Najafi, M., Mortezaee, K., & Majidpoor, J. (2019). Cancer stem cell (CSC) Punfa, W., Yodkeeree, S., Pitchakarn, P., Ampasavate, C., & Limtrakul, P.
resistance drivers. Life Sciences, 234, 116781. https://doi.org/10. (2012). Enhancement of cellular uptake and cytotoxicity of curcumin-
1016/j.lfs.2019.116781 loaded PLGA nanoparticles by conjugation with anti-P-glycoprotein in
Nami, B., & Wang, Z. (2018). Genetics and expression profile of the tubulin drug resistance cancer cells. Acta Pharmacologica Sinica, 33, 823–831.
gene superfamily in breast cancer subtypes and its relation to taxane https://doi.org/10.1038/aps.2012.34
resistance. Cancers (Basel), 10, 274. https://doi.org/10.3390/ Rahmani, A. H., Al Zohairy, M. A., Aly, S. M., & Khan, M. A. (2014). Cur-
cancers10080274 cumin: A potential candidate in prevention of cancer via modulation of
Nikolaou, M., Pavlopoulou, A., Georgakilas, A. G., & Kyrodimos, E. (2018). molecular pathways. BioMed Research International, 2014, 761608.
The challenge of drug resistance in cancer treatment: A current over- https://doi.org/10.1155/2014/761608
view. Clinical & Experimental Metastasis, 35, 309–318. https://doi.org/ Rai, M., Pandit, R., Gaikwad, S., Yadav, A., & Gade, A. (2015). Potential
10.1007/s10585-018-9903-0 applications of curcumin and curcumin nanoparticles: From traditional
Nishinaka, T., Ichijo, Y., Ito, M., Kimura, M., Katsuyama, M., Iwata, K., … therapeutics to modern nanomedicine. Nanotechnology Reviews, 4.
Yabe-Nishimura, C. (2007). Curcumin activates human glutathione S- https://doi.org/10.1515/ntrev-2015-0001
transferase P1 expression through antioxidant response element. Toxi- Ramachandran, C., Rodriguez, S., Ramachandran, R., Nair, P. K. R.,
cology Letters, 170, 238–247. https://doi.org/10.1016/j.toxlet.2007. Fonseca, H., Khatib, Z., … Melnick, S. J. (2005). Expression profiles of
03.011 apoptotic genes induced by curcumin in human breast cancer and
Nobili, S., Lippi, D., Witort, E., Donnini, M., Bausi, L., Mini, E., & mammary epithelial cell lines. Anticancer Research, 25, 3293–3302.
Capaccioli, S. (2009). Natural compounds for cancer treatment and Razaghi, A., Heimann, K., Schaeffer, P. M., & Gibson, S. B. (2018). Negative
prevention. Pharmacological Research, 59, 365–378. https://doi.org/ regulators of cell death pathways in cancer: Perspective on biomarkers
10.1016/j.phrs.2009.01.017 and targeted therapies. Apoptosis, 23, 93–112. https://doi.org/10.
Odot, J., Albert, P., Carlier, A., Tarpin, M., Devy, J., & Madoulet, C. (2004). 1007/s10495-018-1440-4
In vitro and in vivo anti-tumoral effect of curcumin against melanoma Reuter, S., Gupta, S. C., Park, B., Goel, A., & Aggarwal, B. B. (2011). Epige-
cells. International Journal of Cancer, 111, 381–387. https://doi.org/10. netic changes induced by curcumin and other natural compounds.
1002/ijc.20160 Genes & Nutrition, 6, 93–108. https://doi.org/10.1007/s12263-011-
Ooko, E., Alsalim, T., Saeed, B., Saeed, M. E. M., Kadioglu, O., 0222-1
Abbo, H. S., … Efferth, T. (2016). Modulation of P-glycoprotein Revalde, J. L., Li, Y., Hawkins, B. C., Rosengren, R. J., & Paxton, J. W.
activity by novel synthetic curcumin derivatives in sensitive and (2015). Heterocyclic cyclohexanone monocarbonyl analogs of cur-
multidrug-resistant T-cell acute lymphoblastic leukemia cell lines. cumin can inhibit the activity of ATP-binding cassette transporters in
Toxicology and Applied Pharmacology, 305, 216–233. https://doi. cancer multidrug resistance. Biochemical Pharmacology, 93, 305–317.
org/10.1016/j.taap.2016.06.002 https://doi.org/10.1016/j.bcp.2014.12.012
Palumbo, M. O., Kavan, P., Miller, W. H., Panasci, L., Assouline, S., Rives, V., Del Arco, M., & Martín, C. (2013). Layered double hydroxides as
Johnson, N., … Batist, G. (2013). Systemic cancer therapy: Achieve- drug carriers and for controlled release of non-steroidal
ments and challenges that lie ahead. Frontiers in Pharmacology, 4, 57. antiinflammatory drugs (NSAIDs): A review. Journal of Controlled
https://doi.org/10.3389/fphar.2013.00057 Release, 169, 28–39. https://doi.org/10.1016/j.jconrel.2013.03.034
Pan, S.-T., Li, Z.-L., He, Z.-X., Qiu, J.-X., & Zhou, S.-F. (2016). Molecular Rivlin, N., Brosh, R., Oren, M., & Rotter, V. (2011). Mutations in the p53
mechanisms for tumour resistance to chemotherapy. Clinical and tumor suppressor gene: Important milestones at the various steps of
Experimental Pharmacology & Physiology, 43, 723–737. https://doi.org/ tumorigenesis. Genes and Cancer, 2, 466–474. https://doi.org/10.
10.1111/1440-1681.12581 1177/1947601911408889
Panda, A. K., Chakraborty, D., Sarkar, I., Khan, T., & Sa, G. (2017). New Robey, R. W., Polgar, O., Deeken, J., To, K. W., & Bates, S. E. (2007).
insights into therapeutic activity and anticancer properties of cur- ABCG2: determining its relevance in clinical drug resistance. Cancer
cumin. Journal of Experimental Pharmacology, 9, 31–45. https://doi. and Metastasis Reviews, 26, 39–57. https://doi.org/10.1007/s10555-
org/10.2147/JEP.S70568 007-9042-6
Park, J. (2010). Anti-carcinogenic properties of curcumin on colorectal can- Rocha, C., Silva, M., Quinet, A., Cabral-Neto, J., & Menck, C. (2018). DNA
cer. World Journal of Gastrointestinal Oncology, 2, 169–176. https:// repair pathways and cisplatin resistance: An intimate relationship.
doi.org/10.4251/wjgo.v2.i4.169 Clinics, 73, 1–10. https://doi.org/10.6061/clinics/2018/e478s
Pathania, S., Bhatia, R., Baldi, A., Singh, R., & Rawal, R. K. (2018). Drug Rowe, D. L., Ozbay, T., O'Regan, R. M., & Nahta, R. (2009). Modulation of
metabolizing enzymes and their inhibitors' role in cancer resistance. the BRCA1 protein and induction of apoptosis in triple negative breast
Biomedicine & Pharmacotherapy, 105, 53–65. https://doi.org/10.1016/ cancer cell lines by the polyphenolic compound curcumin. Breast Can-
j.biopha.2018.05.117 cer: Basic and Clinical Research, 3, 61–75.
Peng, X. X., Tiwari, A. K., Wu, H. C., & Chen, Z. S. (2012). Overexpression Roy, M., & Mukherjee, S. (2014). Reversal of resistance towards cisplatin by
of P-glycoprotein induces acquired resistance to imatinib in chronic curcumin in cervical cancer cells. Asian Pacific Journal of Cancer Prevention,
myelogenous leukemia cells. Chinese Journal of Cancer, 31, 110–118. 15, 1403–1410. https://doi.org/10.7314/apjcp.2014.15.3.1403
https://doi.org/10.5732/cjc.011.10327 Roy, S., Yu, Y., Padhye, S. B., Sarkar, F. H., & Majumdar, A. P. N. (2013).
Pfeffer, C. M., & Singh, A. T. K. (2018). Apoptosis: A target for anticancer Difluorinated-curcumin (CDF) restores PTEN expression in colon can-
therapy. International Journal of Molecular Sciences, 19, 448. https:// cer cells by down-regulating miR-21. PLoS ONE, 8, 5–10. https://doi.
doi.org/10.3390/ijms19020448 org/10.1371/journal.pone.0068543
Pommier, Y. (2013). Drugging topoisomerases: Lessons and challenges. Saha, S., Adhikary, A., Bhattacharyya, P., Das, T., & Sa, G. (2012). Death by
ACS Chemical Biology, 8, 82–95. https://doi.org/10.1021/cb300648v design: Where curcumin sensitizes drug-resistant tumours. Anticancer
Pongrakhananon, V., & Rojanasakul, Y. (2012). Anticancer properties of Research, 32, 2567–2584.
curcumin. Advanced Cancer Therapeutics, 345–368. https://doi.org/10. Salehan, M. R., & Morse, H. R. (2013). DNA damage repair and tolerance:
5772/23594 A role in chemotherapeutic drug resistance. British Journal of Biomedi-
Prieto-Vila, M., Takahashi, R. U., Usuba, W., Kohama, I., & Ochiya, T. cal Science, 70, 31–40. https://doi.org/10.1080/09674845.2013.
(2017). Drug resistance driven by cancer stem cells and their niche. 11669927
International Journal of Molecular Sciences, 18, 2574. https://doi.org/ Sanaei, M., & Kavoosi, F. (2018). Effect of curcumin and trichostatin a on
10.3390/ijms18122574 the expression of DNA methyltransfrase 1 in hepatocellular carcinoma
22 KEYVANI-GHAMSARI ET AL.

cell line hepa 1-6. The Iranian Journal of Pediatric Hematology & Oncol- therapies. Clinical Cancer Research, 22, 5651–5660. https://doi.org/10.
ogy, 8, 193–201. 1158/1078-0432.CCR-16-0247
Sarkadi, B., Homolya, L., Szakács, G., & Váradi, A. (2006). Human multidrug Subramaniam, D., Ponnurangam, S., Ramamoorthy, P., Standing, D.,
resistance ABCB and ABCG transporters: Participation in a Battafarano, R. J., Anant, S., & Sharma, P. (2012). Curcumin induces
chemoimmunity defense system. Physiological Reviews, 86, cell death in esophageal cancer cells through modulating notch signal-
1179–1236. https://doi.org/10.1152/physrev.00037.2005 ing. PLoS ONE, 7, e30590. https://doi.org/10.1371/journal.pone.
Selvendiran, K., Tong, L., Vishwanath, S., Bratasz, A., Trigg, N. J., 0030590
Kutala, V. K., … Kuppusamy, P. (2007). EF24 induces G2/M arrest and Syng-Ai, C., Kumari, A. L., & Khar, A. (2004). Effect of curcumin on normal
apoptosis in cisplatin-resistant human ovarian cancer cells by increas- and tumor cells: Role of glutathione and bcl-2. Molecular Cancer Thera-
ing PTEN expression. The Journal of Biological Chemistry, 282, peutics, 3, 1101–1108.
28609–28618. https://doi.org/10.1074/jbc.M703796200 Talalay, P. (2000). Chemoprotection against cancer by induction of phase
Shakibaei, M., Buhrmann, C., Kraehe, P., Shayan, P., Lueders, C., & Goel, A. 2 enzymes. BioFactors, 12, 5–11. https://doi.org/10.1002/biof.
(2014). Curcumin chemosensitizes 5-fluorouracil resistant MMR- 5520120102
deficient human colon cancer cells in high density cultures. PLoS ONE, Talib, W. H., Al-hadid, S. A., Wild Ali, M. B., Al-Yasari, I. H., & Abd
9, 1–12. https://doi.org/10.1371/journal.pone.0085397 Ali, M. R. (2018). Role of curcumin in regulating p53 in breast
Shang, H. S., Chang, C. H., Chou, Y. R., Yeh, M. Y., Au, M. K., Lu, H. F., … cancer: An overview of the mechanism of action. Breast Cancer: Tar-
Chung, J. G. (2016). Curcumin causes DNA damage and affects gets and Therapy, 10, 207–217. https://doi.org/10.2147/BCTT.
associated protein expression in HeLa human cervical cancer cells. S167812
Oncology Reports, 36, 2207–2215. https://doi.org/10.3892/or. Tan, B. L., & Norhaizan, M. E. (2019). Curcumin combination chemother-
2016.5002 apy: The implication and efficacy in cancer. Molecules, 24, 1–21.
Shanmugam, M. K., Rane, G., Kanchi, M. M., Arfuso, F., Chinnathambi, A., https://doi.org/10.3390/molecules24142527
Zayed, M. E., … Sethi, G. (2015). The multifaceted role of curcumin in Tomeh, M. A., Hadianamrei, R., & Zhao, X. (2019). A review of curcumin
cancer prevention and treatment. Molecules, 20, 2728–2769. https:// and its derivatives as anticancer agents. International Journal of Molecu-
doi.org/10.3390/molecules20022728 lar Sciences, 20, 1033. https://doi.org/10.3390/ijms20051033
Sharma, R. A., Euden, S. A., Platton, S. L., Cooke, D. N., Shafayat, A., Tripathi, A., & Misra, K. (2016). Designing and development of novel cur-
Hewitt, H. R., … Steward, W. P. (2004). Phase I clinical trial of oral cur- cumin analogues/congeners as inhibitors of breast Cancer stem cells
cumin: Biomarkers of systemic activity and compliance. Clinical Cancer growth. Chemical Engineering Transactions, 49, 79–84. https://doi.org/
Research, 10, 6847–6854. https://doi.org/10.1158/1078-0432.CCR- 10.3303/CET1649014
04-0744 Tsai, L. L., Yu, C. C., Lo, J. F., Sung, W. W., Lee, H., Chen, S. L., &
Shimizu, M., Gruz, P., Kamiya, H., Kim, S. R., Pisani, F. M., Masutani, C., … Chou, M. Y. (2012). Enhanced cisplatin resistance in oral-cancer stem-
Nohmi, T. (2003). Erroneous incorporation of oxidized DNA precursors like cells is correlated with upregulation of excision-repair cross-
by Y-family DNA polymerases. EMBO Reports, 4, 269–273. https:// complementation group 1. The Journal of Dental Sciences, 7, 111–117.
doi.org/10.1038/sj.embor.embor765 https://doi.org/10.1016/j.jds.2012.03.006
Shishodia, S., Amin, H. M., Lai, R., & Aggarwal, B. B. (2005). Curcumin Um, Y., Cho, S., Woo, H. B., Kim, Y. K., Kim, H., Ham, J., … Lee, S. (2008).
(diferuloylmethane) inhibits constitutive NF-κB activation, induces Synthesis of curcumin mimics with multidrug resistance reversal activi-
G1/S arrest, suppresses proliferation, and induces apoptosis in mantle ties. Bioorganic & Medicinal Chemistry, 16, 3608–3615. https://doi.org/
cell lymphoma. Biochemical Pharmacology, 70, 700–713. https://doi. 10.1016/j.bmc.2008.02.012
org/10.1016/j.bcp.2005.04.043 Vallianou, N. G., Evangelopoulos, A., Schizas, N., & Kazazis, C. (2015).
Siddik, Z. H. (2003). Cisplatin: Mode of cytotoxic action and molecular Potential anticancer properties and mechanisms of action of curcumin.
basis of resistance. Oncogene, 22, 7265–7279. https://doi.org/10. Anticancer Research, 35, 645–651.
1038/sj.onc.1206933 Vinod, B. S., Antony, J., Nair, H. H., Puliyappadamba, V. T., Saikia, M., Shyam
Simental-Mendía, L. E., & Sahebkar, A. (2016). Modulation of microRNAs Narayanan, S., … John Anto, R. (2013). Mechanistic evaluation of the sig-
by curcumin in pancreatic cancer. Clinical Nutrition, 35, 1585. https:// naling events regulating curcumin-mediated chemosensitization of breast
doi.org/10.1016/j.clnu.2016.09.001 cancer cells to 5-fluorouracil. Cell Death & Disease, 4, e505–e513.
Soria, J. C., Smit, E., Khayat, D., Besse, B., Yang, X., Hsu, C. P., … https://doi.org/10.1038/cddis.2013.26
Blackhall, F. (2010). Phase 1b study of dulanermin (recombinant Vosooghi, M., Firoozpour, L., Rodaki, A., Pordeli, M., Safavi, M.,
human Apo2L/TRAIL) in combination with paclitaxel, carboplatin, and Ardestani, S. K., … Foroumadi, A. (2014). Design, synthesis, docking
bevacizumab in patients with advanced non-squamous non-small-cell study and cytotoxic activity evaluation of some novel letrozole ana-
lung cancer. Journal of Clinical Oncology, 28, 1527–1533. https://doi. logs. DARU. Journal of Pharmaceutical Sciences, 22, 1–7. https://doi.
org/10.1200/JCO.2009.25.4847 org/10.1186/s40199-014-0083-4
Spiros, A. V. (2017). Aberrant control of NF-κB in cancer permits transcrip- Wang, B., Liu, X., Teng, Y., Yu, T., Chen, J., Hu, Y., … Shen, Y. (2017).
tional and phenotypic plasticity, to curtail dependence on host tissue: Improving anti-melanoma effect of curcumin by biodegradable
Molecular mode. Cancer Biology & Medicine, 14, 254. https://doi.org/ nanoparticles. Oncotarget, 8, 108624–108642. https://doi.org/10.
10.20892/j.issn.2095-3941.2017.0029 18632/oncotarget.20585
Sreenivasan, S., Thirumalai, K., Danda, R., & Krishnakumar, S. (2012). Effect Wang, P., Yang, H. L., Yang, Y. J., Wang, L., & Lee, S. C. (2015). Overcome
of curcumin on miRNA expression in human Y79 retinoblastoma cells. Cancer Cell Drug Resistance Using Natural Products. Evidence-Based
Current Eye Research, 37, 421–428. https://doi.org/10.3109/ Complementary and Alternative Medicine, 2015, 14. https://doi.org/10.
02713683.2011.647224 1155/2015/767136
Srivastava, A. K., Han, C., Zhao, R., Cui, T., Dai, Y., Mao, C., … Wang, Q.-E. Wang, Z., Sun, H., Provaznik, J., Hackert, T., & Zöller, M. (2019). Pancreatic
(2015). Enhanced expression of DNA polymerase eta contributes to cancer-initiating cell exosome message transfer into noncancer-
cisplatin resistance of ovarian cancer stem cells. Proceedings of the initiating cells: The importance of CD44v6 in reprogramming. Journal
National Academy of Sciences, 112, 4411–4416. https://doi.org/10. of Experimental & Clinical Cancer Research, 38, 1–20. https://doi.org/
1073/pnas.1421365112 10.1186/s13046-019-1129-8
Stover, E. H., Konstantinopoulos, P. A., Matulonis, U. A., & Swisher, E. M. Wang, Z., Sun, W., Huang, C. K., Wang, L., Ia, M. M., Cui, X., … Wang, Z. S.
(2016). Biomarkers of response and resistance to DNA repair targeted (2015). Inhibitory effects of curcumin on activity of cytochrome P450
KEYVANI-GHAMSARI ET AL. 23

2C9 enzyme in human and 2C11 in rat liver microsomes. Drug Devel- through influencing Cu-Sp1-CTR1 regulatory loop. Phytomedicine, 48,
opment and Industrial Pharmacy, 41, 613–616. https://doi.org/10. 51–61. https://doi.org/10.1016/j.phymed.2018.04.058
3109/03639045.2014.886697 Zhang, X., Chen, Q., Wang, Y., Peng, W., & Cai, H. (2014). Effects of cur-
Wei, Y., Pu, X., & Zhao, L. (2017). Preclinical studies for the combina- cumin on ion channels and transporters. Frontiers in Pharmacology, 5,
tion of paclitaxel and curcumin in cancer therapy (review). Oncol- 1–6. https://doi.org/10.3389/fphys.2014.00094
ogy Reports, 37, 3159–3166. https://doi.org/10.3892/or.2017. Zhang, Y., & Wang, J. (2017). MicroRNAs are important regulators of drug
5593 resistance in colorectal cancer. Biological Chemistry, 398, 929–938.
Wilken, R., Veena, M. S., Wang, M. B., & Srivatsan, E. S. (2011). Curcumin: https://doi.org/10.1515/hsz-2016-0308
A review of anti-cancer properties and therapeutic activity in head and Zhang, Z., Han, H., Rong, Y., Zhu, K., Zhu, Z., Tang, Z., … Tao, J. (2018).
neck squamous cell carcinoma. Molecular Cancer, 10, 12. https://doi. Hypoxia potentiates gemcitabine-induced stemness in pancreatic
org/10.1186/1476-4598-10-12 cancer cells through AKT/Notch1 signaling. Journal of Experimental &
Wilkens, S. (2015). Structure and mechanism of ABC transporters. Clinical Cancer Research, 37, 1–13. https://doi.org/10.1186/
F1000Prime Reports, 7, 14. https://doi.org/10.12703/P7-14 s13046-018-0972-3
Wong, R. S. Y. (2011). Apoptosis in cancer: From pathogenesis to treat- Zhao, G., Han, X., Zheng, S., Li, Z., Sha, Y., Ni, J., … Song, Z. (2016). Curcumin
ment. Journal of Experimental & Clinical Cancer Research, 30, 87. induces autophagy, inhibits proliferation and invasion by downregulating
https://doi.org/10.1186/1756-9966-30-87 AKT/mTOR signaling pathway in human melanoma cells. Oncology
Xia, L., Tan, S., Zhou, Y., Lin, J., Wang, H., Oyang, L., … Liao, Q. (2018). Role Reports, 35, 1065–1074. https://doi.org/10.3892/or.2015.4413
of the NFκB-signaling pathway in cancer. Oncotargets and Therapy, 11, Zhao, Q., Guan, J., Qin, Y., Ren, P., Zhang, Z., Lv, J., … Mao, W. (2018). Cur-
2063–2073. https://doi.org/10.2147/OTT.S161109 cumin sensitizes lymphoma cells to DNA damage agents through regu-
Yan, G., Graham, K., & Lanza-Jacoby, S. (2013). Curcumin enhances the lating Rad51-dependent homologous recombination. Biomedicine &
anticancer effects of trichostatin a in breast cancer cells. Molecular Pharmacotherapy, 97, 115–119. https://doi.org/10.1016/j.biopha.
Carcinogenesis, 52, 404–411. https://doi.org/10.1002/mc.21875 2017.09.078
Yang, J., Cao, Y., Sun, J., & Zhang, Y. (2010). Curcumin reduces the expres- Zheng, J., Wu, C., Lin, Z., Guo, Y., Shi, L., Dong, P., … Yu, F. (2014). Cur-
sion of Bcl-2 by upregulating miR-15a and miR-16 in MCF-7 cells. cumin up-regulates phosphatase and tensin homologue deleted on
Medical Oncology, 27, 1114–1118. https://doi.org/10.1007/s12032- chromosome 10 through microRNA-mediated control of DNA
009-9344-3 methylation—A novel mechanism suppressing liver fibrosis. The FEBS
Yallapu, M. M., Jaggi, M., & Chauhan, S. C. (2013). Curcumin nanomedicine: A Journal, 281, 88–103. https://doi.org/10.1111/febs.12574
road to cancer therapeutics. Current Pharmaceutical Design, 19, Zhou, W., Chen, Y. W., Liu, X., Chu, P., Loria, S., Wang, Y., … Chou, K. M.
1994–2010. https://doi.org/10.2174/1381612811319110004 (2013). Expression of DNA translesion synthesis polymerase η in head
You, Y., Bi, F. F., Jiang, Y., Xu, Y. T., An, Y. Y., Li, D., & Yang, Q. (2019). and neck squamous cell cancer predicts resistance to gemcitabine and
BRCA1 affects the resistance and stemness of SKOV3-derived ovarian cisplatin-based chemotherapy. PLoS ONE, 8, 1–10. https://doi.org/10.
cancer stem cells by regulating autophagy. Cancer Medicine, 8, 1371/journal.pone.0083978
656–668. https://doi.org/10.1002/cam4.1975 Zou, J., Zhu, L., Jiang, X., Wang, Y., Wang, Y., Wang, X., & Chen, B. (2018).
Zahreddine, H., & Borden, K. L. B. (2013). Mechanisms and insights into Curcumin increases breast cancer cell sensitivity to cisplatin by
drug resistance in cancer. Frontiers in Pharmacology, 4, 1–8. https:// decreasing FEN1 expression. Oncotarget, 9, 11268–11278. https://
doi.org/10.3389/fphar.2013.00028 doi.org/10.18632/oncotarget.24109
Zaman, M. S., Chauhan, N., Yallapu, M. M., Gara, R. K., Maher, D. M.,
Kumari, S., … Chauhan, S. C. (2016). Curcumin nanoformulation for
cervical cancer treatment. Scientific Reports, 6, 1–14. https://doi.org/
10.1038/srep20051
Zhang, C., Xu, Y., Wang, H., Li, G., Yan, H., Fei, Z., … Li, W. (2018). Cur- How to cite this article: Keyvani-Ghamsari S, Khorsandi K,
cumin reverses irinotecan resistance in colon cancer cell by regulation Gul A. Curcumin effect on cancer cells' multidrug resistance:
of epithelial-mesenchymal transition. Anti-Cancer Drugs, 29, 334–340. An update. Phytotherapy Research. 2020;1–23. https://doi.
https://doi.org/10.1097/CAD.0000000000000599
org/10.1002/ptr.6703
Zhang, W., Shi, H., Chen, C., Ren, K., Xu, Y., Liu, X., & He, L. (2018). Cur-
cumin enhances cisplatin sensitivity of human NSCLC cell lines

You might also like